Abstract
Free full text
The classification of microglial activation phenotypes on neurodegeneration and regeneration in Alzheimer’s disease brain
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disease characterized by progressive decline of cognitive function and memory formation. There is no therapeutic that can halt or reverse its progression. Contemporary research suggests that age-dependent neuroinflammatory changes may play a significant role in the decreased neurogenesis and cognitive impairments in AD. The innate immune response is characterized by pro-inflammatory (M1) activation of macrophages and subsequent production of specific cytokines, chemokines, and reactive intermediates, followed by resolution and alternative activation for anti-inflammatory signaling (M2a) and wound healing (M2c). We propose that microglial activation phenotypes are analogous to those of macrophages and that their activation plays a significant role in regulating neurogenesis in the brain. Microglia undergo a switch from an M2- to an M1-skewed activation phenotype during aging. This review will assess the neuroimmunological studies that led to characterization of the different microglial activation states using AD mouse models. It will also discuss the roles of microglial activation on neurogenesis in AD and propose anti-inflammatory molecules as exciting therapeutic targets for research. Molecules like interleukin-4 and CD200 have proven to be important anti-inflammatory molecules in the regulation of neuroinflammation in the brain, and they will be discussed in detail for their therapeutic potential.
1. Introduction
Alzheimer’s disease (AD) is the most prevalent neurodegenerative disease, and it is characterized by progressive decline of cognitive function and memory formation in mostly elderly populations (Minati et al., 2009). While the exact pathophysiology of AD has yet to be determined, it is known that the greatest risk factor for development of the disease is age. AD presents with a very characteristic neuropathology: intraneuronal accumulations of hyperphosphorylated microtuble-associated protein tau known as neurofibrillary tangles (NFTs), and extracellular deposition of amyloid β-peptide (Aβ) known as amyloid or senile plaques (Dickson et al., 1988; Alzheimer et al., 1995). AD patients also exhibit a significant loss of synapses and neurons, namely in the temporal and frontal cortices and hippocampi (Davies et al., 1987; Scheff et al., 2006; Scheff et al., 2007). Increasing evidence suggests that it is the aggregation of Aβ fragments that causes neurotoxic effects that lead to the neural, synaptic, and cognitive degradation seen in AD, making Aβ the target of most AD research today (Hardy and Selkoe, 2002).
2. APP and Aβ biology
Aβ is formed by the proteolytic cleavage of amyloid precursor protein (APP) (Zhao et al.), a protein that contains an Aβ-encoding region. As APP is secreted from the cell, it undergoes two consecutive proteolytic cleavages by a series of secretases: α then γ, or β then γ (Esch et al., 1990). Members of the ADAM (a disintegrin and metalloproteinase) family have α-secretase activity and are implicated in APP cleavage (Zhang et al., 2011), but because α-secretase cleaves within the Aβ encoding region, it is not a factor in Aβ formation. β-site APP converting enzyme (BACE) 1 and 2 are β-secretases, and both have been implicated as major factors in the formation of Aβ (Vassar et al., 1999). The γ-secretase complex consists of four components: presenilin 1 or 2 (PS1, PS2, the catalytic domains), Nicastrin, APH-1, and presenilin enhancer-2 (PEN-2) (Kimberly et al., 2003; Takasugi et al., 2003). Missense mutations within the PS1 and PS2 genes are the most common cause of early onset familial AD (Levy-Lahad et al., 1995; Sherrington et al., 1995), but mutations in APP have also been seen in more elderly familial cases (Citron et al., 1992). In addition to cleaving APP, γ-secretase also cleaves a large number of transmembrane proteins, including Notch (Haass and De Strooper, 1999), whose cleaved intracellular domain plays an important role as a transcriptional factor in regulating genes involved in development (Kopan et al., 1996; Schroeter et al., 1998).
For non-amyloidogenic cleavage of APP, α-secretase cleaves the protein between amino acid residues 16 and 17 within the Aβ region, creating a short 83-residue fragment known as p83 (Esch, et al. 1990). The p83 fragment is subsequently cleaved by γ-secretase within the transmembrane region, generating p3 (Aβ17-40) and APP intracellular domain (AICD). AICD can potentially translocate to the nucleus and function as a transcription factor Cao and Sudhof (2001); (Baek et al., 2002; Leissring et al., 2002; Cao and Sudhof, 2004). The Aβ fragment is created by cleavage with β- then γ-secretases (Seubert et al., 1993). β-secretase cleaves at the N-terminus of the Aβ region and is followed by cleavage by γ-secretase to create a 40-42-residue Aβ, with the majority of Aβ fragments being Aβ40, and the less common, more amyloidogenic species being Aβ42. Studies of Aβ deposition in Down Syndrome and biochemical studies of both Aβ40 and Aβ42 have shown that the Aβ42 fragment tends to aggregate into fibrils more readily than the Aβ40 fragment, causing a gradual increase of Aβ42 deposition with age (Jarrett et al., 1993; Iwatsubo et al., 1995; Lemere et al., 1996).
A large number of studies have proven that Aβ has a neurotoxic effect when aggregated in specific forms (Lambert et al., 1998; Hardy and Selkoe, 2002; Walsh et al., 2002). Its formation and build-up precedes the accumulation of hyperphosphorylated tau into NFTs and leads to synaptic dysfunction and neuronal loss (Selkoe, 1991; Mattson et al., 1998; Lue et al., 1999; Shankar and Walsh, 2009). These findings, together with the fact that select familial AD is due to causative gene mutations in the coding sequences of APP, PS1 and PS2, which are directly involved in Aβ formation, led to the development of the “Amyloid Cascade Hypothesis” first proposed by Selkoe in 1996 (Selkoe, 1996). The Amyloid Cascade Hypothesis states that abnormal processing of APP, whether genetic or spontaneous, causes overproduction of Aβ42 fragments. The brain is unable to clear the Aβ, and it gradually accumulates within the brain. The Aβ aggregates are toxic to neurons and synapses, and this leads to innate immune activation of microglia and astrocytes and attendant inflammatory processes, although the exact pathophysiology of this step is largely unknown (McGeer and McGeer, 2010). There is subsequent production and build-up of reactive intermediates and oxygen species that lead to oxidative injury (Kish et al., 1992; Gutowicz, 2011). Other changes in kinase and phosphatase activities lead to the formation of NFTs (Iqbal et al., 1998). These combined toxic effects are thought to be the underlying cause of neuronal loss, synaptic dysfunction, and the subsequent degradation of cognitive function in AD, and this is currently the most widely accepted hypothesis.
The Amyloid Cascade Hypothesis, however, cannot completely explain the degradation in AD based on several pieces of evidence (Struble et al., 2010). Firstly, therapeutics against Aβ, though successful in removing Aβ fragments in animal models and humans, do not stop the progression of AD, meaning it is not the single cause of dementia (Holmes et al., 2008; Salloway et al., 2009). Secondly, Aβ is normally found even in the healthy human brain and in normal cell culture of healthy humans and other mammals (Seubert et al., 1992; Shoji et al., 1992; Haass and De Strooper, 1999), and those individuals with APP or γ-secretase mutations that have higher deposits of Aβ do not show dementia until they are 20-40 years old (Bird, 2008). In fact, about 20-30% of individuals with normal cognition who never develop dementia show significant levels of amyloid burden in the brain (Bennett et al., 2006; Aizenstein et al., 2008). This has led research to aim for the discovery of the other underlying factor(s) that leads to the cognitive decline in AD.
3. Neuroinflammation
It has long been known that inflammation is a prominent factor, not just in AD, but in a number of age-associated diseases, including atherosclerosis, diabetes, rheumatoid arthritis, and multiple sclerosis (MS). In fact, data from transcriptome analyses of brain tissue from normally aging humans and mice showed significant up-regulation of certain genes that occurred with age, and about half of those genes were associated with inflammation and oxidative stress (Prolla, 2002; Lu et al., 2004; Loerch et al., 2008; Bishop et al., 2010). This can be partly corrected by caloric restriction (Lee et al., 2000), which suggests that energy expenditure-associated oxidative stress underlies the M1 to M2 conversion in the brain. The outcome of oxidative stress can be DNA damage and reduced repair (Haigis and Yankner, 2010). Highly conserved insulin/IGF-1, TOR, and sirtuin signaling pathways regulate these cellular responses, and the aging process potentially induces their dysregulation, leading to pro-inflammatory microglial activation.
Several pieces of evidence indicate the increased presence of inflammatory molecules in AD, including some of the following: the presence of elevated levels of pro-inflammatory cytokines like tumor necrosis factor-α (TNF-α) in the serum of AD patients (Fillit et al., 1991) and interleukin (IL) -6 in brain tissue (Bauer et al., 1991; Bauer et al., 1992) compared to age-matched controls; the increase of activated microglial cells surrounding senile plaques in the cerebral cortex (Haga et al., 1989); the localization of Aβ deposits to T cells, activated microglia, and reactive astrocytes in AD brains (Wegiel and Wisniewski, 1990; Wisniewski and Wegiel, 1991; Ishii and Haga, 1992); and of particular interest in this article is the down-regulated expression of a neuroimmune regulatory protein, cluster of differentiation 200 (CD200), seen in post-mortem brains of AD patients (Walker et al., 2009).
These findings provide evidence for the new hypothesis that the neuronal damage seen in AD is not simply due to the Aβ deposition or intracellular tau accumulation, but to the brain’s dysregulated inflammatory and oxidative responses to them (McGeer et al., 1989; McGeer et al., 1994). Indeed, some of the people with increased Aβ load did not develop AD, suggesting that a second factor is necessary for the disease development, such as microgliosis. Using two carbon 11 ([11C])-labeled positron emission tomographic imaging agents, Pittsburgh Compound B (PIB, an Aβ marker) and PK-11195 (a benzodiazepine receptor ligand and a microglial marker), several groups examined if PIB and PK11195 binding correlated with cognitive function of AD patients. Two studies reported a negative correlation of cognitive function with microgliosis (PK-11195) but not with Aβ load (PIB) (Edison et al., 2008; Yokokura et al., 2011). However, another small-scale study showed no correlation of PK-11195 binding with clinical diagnosis (Wiley et al., 2009). Other studies showed that, although PIB binding was positively correlated with astrogliosis, it was largely independent from PK-11195 binding (Okello et al., 2009; Kadir et al., 2011). These studies suggest that Aβ load is directly correlated with astrogliosis but not with microgliosis, and therefore, microgliosis is potentially more associated with the cognitive decline in AD.
Of particular interest is the role of the microglial activation switch from the alternative to classical activation phenotype. This change in microglial activation skewing phenotype may be the cause of significant amounts of cortical and hippocampal atrophy that makes neurogenesis a prime target for studying ways to replenish that loss (Voloboueva and Giffard, 2011). This in turn draws attention to the increasing significance of the role of immunomodulatory molecules, such as CD200, in neuroimmune regulation. This review will go over the innate and adaptive immune systems of the brain and the role of classically or alternatively activated microglia, which lead into the possible mechanisms by which neurogenesis can be restored in those areas most affected by neuroinflammation. Lastly, this review will discuss the role of CD200 in immune homeostasis and how it may be used to reverse neuroinflammation based on neuroimmunological studies of AD mouse models and AD patients.
4. The Innate and Adaptive Immune Responses
The immune response of the central nervous system (CNS) follows a well-characterized signaling pathway in response to tissue injury, cell death, and pathogens (Carpentier and Palmer, 2009). The main cells that direct the innate immune response in the CNS are mononuclear phagocytes (brain resident microglia and perivascular macrophages). These cells function as sentinels in the brain and prominently express pathogen recognition receptors (PRRs) that recognize the signaling pathways activated or released by dying or damaged cells known as pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs). PRRs include Toll-like receptors (TLRs), Nucleotide Oligomerization Domain (NOD)-like receptors (NLRs), and Retinoic acid-Inducible Gene 1 (RIG1)-like receptors (RLRs). TLRs are ubiquitously expressed and are often expressed on the plasma membranes of immune cells and glial cells, including microglia, astrocytes, and neurons (Bsibsi et al., 2002; Tang et al., 2007). Interestingly, Aβ itself also acts as a PAMP molecule and interacts with TLRs (Lotz et al., 2005; Reed-Geaghan et al., 2009). NLRs are found in the cytoplasm, and some NOD ligands, such as muramyl dipeptide (MDP), are also expressed in microglia, astrocytes, and neurons, and they have been shown to activate the neuronal cells in vitro (Chauhan et al., 2009; Li et al., 2010). When stimulated by a ligand, such as lipopolysaccharide (LPS, a TLR ligand) or MDP, TLRs or NLRs become activated and initiate transcriptional up-regulation and release of pro-inflammatory cytokines and chemokines (Bailey et al., 2006). This LPS-induced inflammatory response is enhanced in aged animals in vivo (Sparkman and Johnson, 2008; Lynch, 2010). The cytokines released include pro-inflammatory cytokines, such as TNF-α, interferon-γ (IFN-γ), IL-6, and IL-1β. The cytokines initiate the adaptive immune response, activate vasculature, and recruit circulating lymphocytes to the infection or injury. The lymphocytes bind to the activated endothelium and diffuse throughout the affected tissue. Through antigen recognition and with the help of B-cell-mediated antibody production, they remove pathogens by T-cell-mediated destruction of cells. Following the elimination of pathogens, the immune response self-limits itself by the clearance of the molecular patterns that initially triggered the immune response and by Fas ligand (FasL)-mediated apoptosis of activated T cells (Niederkorn, 2006). Both T and B cells undergo apoptosis. The hypothalamo-pituitary-adrenal (HPA) axis is activated during inflammation and increases the production of glucocorticoids to conversely decrease pro-inflammatory signaling. Perivascular macrophages are essential for the anti-inflammatory HPA response to IL-1 (Serrats et al., 2010). Without them, CNS responses to inflammatory insults are enhanced. These cells induce the HPA axis and constrain endothelial cell involvement in prostanoid production during the response to pro-inflammatory signals (Serrats et al., 2010). With the help of lymphocytes, microglia, and neurons, the HPA axis also contributes to the production of anti-inflammatory hormones and cytokines such as IL-4, IL-10, and transforming growth factor-β (TGF-β) (Elenkov and Chrousos, 2002). With that, the immune signaling cascade is resolved and returned to its previous state of surveillance of the CNS.
5. Classification of Macrophage Activation – M1/M2 Switch of activation phenotype
5.1. Classical (M1) Activation
Macrophages are a type of white blood cell derived from monocytes that aid in the removal of foreign substances or organisms by phagocytosis (Mackaness, 1970; David, 1975; Oppenheim and Rosenstreich, 1976). Macrophage activation is classified into two phenotypes: classical and alternative (Mills et al., 2000; Gordon, 2003). Classical activation involves the induction of M1 macrophages by Th1 cell-derived cytokines, like IFN-γ, TNF-α, and IL-1β, to allow cells to respond to PAMPs. M1 macrophages express opsonic receptors, such as immunoglobulin Fc γ receptors (e.g. FcγRIII) (Unkeless et al., 1988; Ravetch and Kinet, 1991). Their activation causes the cells to release high levels of pro-inflammatory cytokines, including TNF-α, STAT3, IL-6, IL-12, IL-1β, and IL-23, nitric oxide synthase (NOS), NADPH oxidases (NOX), and corresponding toxic intermediates, and reactive oxygen and nitrogen species (Gutteridge and Halliwell, 1989; Heinrich et al., 1998; Akiyama et al., 2000; Pratico et al., 2001; Pratico and Sung, 2004; Brown, 2007; Benoit et al., 2008; Burton et al., 2011). Conversely, they have low production of anti-inflammatory cytokines. LPS is known to stimulate M1 activation via the TLR4 complex, which then triggers an intracellular signaling cascade that activates NFκB and three types of mitogen-activated protein kinases (MAPKs): extracellular signal-related kinase (Erk), c-Jun N-terminal kinase (JNK), and p38 (Nagai et al., 2002; Takeda and Akira, 2004). Without some sort of modulation, as from negative feedback, these defense processes could be over-stimulated and cause significant damage. Inflammatory cytokines and reactive oxygen and nitrogen species can lead to widespread cell damage (Gutteridge and Halliwell 1989; Pratico et al. 2001; Pratico and Sung 2004). Classical activation also causes the release of proteolytic enzymes that can lead to deterioration of the extracellular matrix (such as metalloproteinases, collagenases, and furin), thus degrading cellular integrity and leading to easier destruction of the cell (Chandler et al., 1995; Maeda and Sobel, 1996; Rosenberg et al., 2001; Rosenberg, 2009; Shiryaev et al., 2009). It makes sense then that prolonged up-regulation of pro-inflammatory cytokines, like LPS, is accompanied by a decrease in synaptic plasticity, as demonstrated by inhibition of long-term potentiation (LTP) both in vitro and in vivo (Lynch, 2010), as well as a deficit in spatial learning (Oitzl et al., 1993; Hein et al., 2010). To ensure there is negative feedback control, macrophages can assume an alternative activation phenotype for inflammation resolution (Colton, 2009).
5.2. Alternative (M2) Activation
Alternative Activation is induced by Th2 cell-derived cytokine activation of M2 macrophages. This activation leads to the enhanced expression of genes meant for inflammation resolution, immunomodulation, homeostasis, scavenging, angiogenesis, and wound healing. These include genes that repair damage to the extracellular matrix, such as arginase 1 (ARG1) (Jimenez et al., 2008; Gomi et al.), mannose receptor (Akiyama et al., 2000), found in inflammatory zone 1 (FIZZ1), and chitinase 3-like 3 (YM1) (Raes et al., 2002; Colton et al., 2006a). They release high levels of anti-inflammatory cytokines like IL-10, IL-4, IL-13, and TGF-β, and low levels of pro-inflammatory cytokines. M2 macrophages are further characterized into sub-classifications, M2a, M2b, and M2c. M2a macrophages are designed for inflammation resolution and phagocytosis and are induced by IL-4, IL-13, YM1, and ARG1. IL-4 is a particularly important cytokine for M2a skewing (Maher et al., 2005; Nolan et al., 2005; Lyons et al., 2007a). Microglia stimulated by IL-4 have a decrease in TNF-α production and an increase in secretion of insulin-like growth factor-1 (IGF-1), and they also induce neurogenesis in vitro (Butovsky et al., 2005; Butovsky et al., 2006b). M2b macrophages clear away reactive oxygen and nitrogen species released during M1 activation, express Il-10 and CCL1 and they are skewed by immune complexes and TLR or IL-1R agonists (Mantovani et al., 2004). M2c macrophages are for the repair and tissue remodeling (wound healing), express CCL16, CCL17, CCL18, CXCL13, CCR2, and CCR5, and are skewed by IL-10 and glucocorticoid hormones (Mantovani et al., 2004).
6. Classification of Microglial Activation
Microglia are members of the monocytic-macrophage family. Like macrophages, microglia adopt similar activation phenotypes in response to CNS insult. Table 1 characterizes microglial activation states M1, M2a, and M2c by denoting their specific ligands and markers and outlining their effects on neurogenesis, synaptic plasticity, and spatial learning based on in vitro and in vivo experiments, which will be discussed below. Our lab recently identified that resting microglia adopted the M2c phenotype (unpublished observation), showing high expression of TGF-β1, extracellular matrix-related genes, and expression of IL-10. Microglial activation phenotypes switch from M2 to M1 during disease progression. Increased expression of pro-inflammatory cytokines and chemokines is likewise accompanied by M1-skewed microglial activation in the human AD brain (Hoozemans et al., 2006). These findings were reproduced in some AD mouse models, such as the APP+PS1 mouse (Jankowsky et al., 2004), which demonstrated that there was a distinctive shift from M2 to M1 microglial activation in the hippocampus of aged rodents (Jimenez et al., 2008). It was found in aged rats, for example, that IFN-γ increased with age, while Il-4 decreased (Nolan et al., 2005; Maher et al., 2006), suggesting an increased classical activation phenotype over alternative with age. This microglial phenotype switch from M2c to M1 was also demonstrated by in vivo intracerebroventricular injection of LPS into mouse brains as determined by changes in the gene expression of M1 markers (CCL5, CCL11, CXCL9, CXCL10, CXCL11, CXCL16, CD16, IL-12, AND IL-23) and M2c markers (Scavenger Receptor A, Scavenger Receptor B, CD14, CCR2, fibrinogen, collagen, and SOCS3) (Kopydlowski et al., 1999; Mantovani et al., 2004; Lund et al., 2006; Qin et al., 2006; Baker et al., 2009; Villeda et al., 2011).
Table 1
Microglial Activation State | |||
---|---|---|---|
Classification | M1 | M2a | M2c |
Phenotype | Classical/pro- inflammatory Activation | Alternative Activation, anti-inflammatory | Deactivation/Wound- Healing |
Ligands | IFN-γ3, TNF-α3, TLR ligand30, 36; Aβ22, 28, 35 | IL-412, 25, 26, 27, 31, IL- 1329 | IL-1012, glucocorticoid hormones27, TGF-β12 |
Markers | NOX4, NOS214, TNF3, IL-63, IL-123, IL-1β3, CCL23, CCL521, CCL1137, STAT35, 16 | ARG113, 18, MR9, 34, FIZZ19, 34, YM19, 34, IGF-16, 7 | SOCS31, 33, MR9, 34, IL-1012, TGF-β12 |
Neurogenic
Effect | Inhibition of NSC differentiation and maturation 2, 10, 11, 17, 29, 38 | Enhanced NSC differentiation6, 7, 19 | Enhanced NSC proliferation20 |
Neuroprotection | Enhanced neurotoxicity, ECM damage 14 | Anti-inflammatory neuroprotection, phagocytosis8 | Deactivation of glial inflammation27 |
Synaptic
Plasticity | CA1 LTP↓ 23 | CA1 LTP↑ 31 | CA1 LTP↑ 24 |
Spatial Learning | MWM↓15, 32 | RAWM↑ 19 | RAWM↑ 20 |
References | 1,Baker et al 2009; 2,Bastos et al. 2008; 3,Benoit et al. 2008; 4,Brown 2007; 5,Burton et al. 2011; 6,Butovsky et al. 2005; 7,Butovsky et al. 2006b; 8,Carpentier and Palmer 2009; 9,Colton et al. 2006; 10,Deng et al. 2010; 11,Ekdahl et al. 2003; 12,Elenkov and Chrousos 2002; 13,Gomi et al. 2011; 14,Gutteridge and Halliwell 1989; 15Hein et al. 2009; 16,Heinrich et al. 1998; 17,Hoehn et al. 2005; 18,Jimenez et al. 2008; 19,Kiyota et al. 2010; 20,Kiyota et al. 2011; 21,Kopydlowski et al 1999; 22,Lotz et al. 2005; 23,Lynch 2010; 24,Lynch et al. 2004; 25,Lyons et al. 2007a; 26,Maher et al. 2005; 27,Mantovani et al. 2004; 28,E.G. McGeer and McGeer 2010; 29,Monje et al. 2003; 30,Nagai et al. 2002; 31,Nolan et al. 2005; 32,Oitzl et al. 1993; 33,Qin et al 2006; 34,Raes et al. 2002; 35,Reed-Geaghan et al. 2009; 36,Takeda and Akira 2004; 37,Villeda et al. 2011; 38,Walter et al. 2011 |
Further evidence from APP mice and human monocyte-derived macrophages and microglia showed that, while an increase in M1 activators, like IFN-γ receptor and TNF-R1, led to neuroinflammation and inhibited clearance of Aβ, an increase in M2a or M2c activators enhanced Aβ clearance (He et al., 2007; Yamamoto et al., 2007; Yamamoto et al., 2008). The same switch was seen in LPS-stimulated primary microglia (Lund et al., 2006). IL-10 is a modulator of M2c microglial activation (Mantovani et al., 2004). It was found that IL-10 mediated anti-inflammatory responses including decreasing glial activation and production of proinflammatory cytokines (Plunkett et al., 2001). With gene delivery of IL-10 into APP+PS1 mice, there was suppression of astro/microgliosis and a restoration of impaired spatial learning and neurogenesis, and while there was no reduction of β-amyloidosis, there was enhanced vascular transport of Aβ (Kiyota et al., 2011). IL-4, on the other hand, is an important modulator of M2a microglial activation (Mantovani et al., 2004). Gene delivery of IL-4 into APP+PS1 mice partially suppressed glial accumulation in the hippocampus, directly enhanced neurogenesis, restored impaired spatial learning, and also reduced Aβ deposition (Kiyota et al., 2010). Gene delivery of either IL-4 or IL-10 also reversed the LPS-induced deficit in LTP in the rat hippocampus (Lynch et al., 2004; Nolan et al., 2005). These findings and more are outlined in Table 2, which briefly summarizes the results of cytokine modulation on neuroinflammatory effects in AD mouse models. As can be seen in Table 2, APP mice encompass both M1 and M2c phenotypes, suggesting that the expression of the APP gene induces the up-regulation of both pro- and anti-inflammatory cytokines. Also note the contradictory findings in NOS2−/− mice. It was previously found that APP mice crossed with a NOS2−/− exhibited an M1 microglial activation phenotype and enhanced β-amyloidosis and impaired spatial/ contextual learning (Benzing et al., 1999; Apelt and Schliebs, 2001; Abbas et al., 2002). In the most recent publication, however, Kummer et al found that APP+PS1 mice crossed with NOS2−/− had decreased β-amyloidosis and improved spatial/ contextual learning compared to APP+PS1 mice (Kummer et al., 2011). This may be due to their use of the APP+PS1 model instead of the APP-only mouse and differences in nitration of Aβ42. These findings demonstrate that there is an important role for M2a and M2c activators in microglial activation and clearance of Aβ in vivo. Although it is well established that there is a large immune response component to AD, it is still not completely understood which activation phenotype affects the onset or progression of the disease and which should be the therapeutic target. The failure of anti-inflammatory drugs, like non-steroidal anti-inflammatory drugs (NSAIDs), to halt the progression of AD drives the need for more research into the link between neuroinflammation and AD (Firuzi and Pratico, 2006; Martin et al., 2008; Cole and Frautschy, 2010). This could be due to the suppression of, not only pro-inflammatory, but also anti-inflammatory activation by endogenous molecules, inactivating the beneficial effect of M2a or M2c-skewed microglial functions. Microglial activation is a prominent feature of AD and serves as the driving force behind inflammation, and has recently been suggested to play a role in both neuroprotection and neurogenesis (Butovsky et al., 2006b; Zhao et al., 2006; Figueiredo et al., 2008; Shein et al., 2008; Xapelli et al., 2008; Gomi et al., 2011), making researchers question the exact nature of its association with AD pathology.
Table 2
Modified Phenotype | Model | Cytokine Modified | Inflammation | Neurogenesis | Spatial/ Contextual Learning | β- amyloidosis | References |
---|---|---|---|---|---|---|---|
APP | ↑ IFN-γ, IL-1β, IL-6, TGF-β, IL-10, IL-12, ↓ IL-4 | ↑ ↓ | ↓ | + | (Abbas et al. 2002; Apelt and Schliebs 2001; Benzing et al. 1999) | ||
APP+PS1 | ↑ TNF-α, IL-6, IL-12, IL-1β, IL-1α | ↓ | ↓ | ++ | (Patel et al. 2005) | ||
3×Tg-AD | ↑ TNF-α | ↓ | ↓ | + | (Janelsins et al. 2005) | ||
M1↓ | APP | DN C3 TG | ↓ Microgliosis | ND | ND | ↑ | (Wyss-Coray et al. 2002) |
M1↑ | APP | CCL2 TG | ↑ Astro/microgliosis | ND | ↓ | ↑ | (Kiyota et al. 2009a; Song et al. 2011) |
M1↓ | APP+PS1 | DN CCL2 GT | ↓ Astro/microgliosis | ND | ↑ | ↓ | (Kiyota et al. 2009b) |
M1 → M2b | APP+PS1 | × CD14−/− | ↓ Microgliosis, FIZZ1, YM1, ↑ TNF-α, IFN-γ, IL-10 | ND | ND | ↓ | (Reed-Geaghan et al. 2010) |
M1↓ | APP | × IFN-γR−/− | ↓ TNF-α, astro/microgliosis | ND | ND | ↓ | (Yamamoto et al. 2007) |
M1↑ | APP | IL-1β TG | ↑ MHCII, astrogliosis | ND | ND | ↓ | (Shaftel et al. 2007) |
M1↑ | APP | × NOS2−/− | ↑ TNF-α, IL-6 | ND | ↓ | ↑ | (Colton et al. 2006; Colton et al. 2008; Wilcock et al. 2008) |
M1↓ | APP+PS1 | × NOS2−/− | ↓ Astro/microgliosis | ND | ↑ | ↓ | (Kummer et al. 2011) |
M1↓ | APP | × TLR-4 DN TG | ↑↓ Microgliosis ↓IL-1β, IL-6 | ND | ↓ | ↑ | (Jin et al. 2008; Tahara et al. 2006) |
M1↓ | APP | × TNFR1−/− | ↓ Astro/microgliosis | ND | ↑ | ↓ | (He et al. 2007) |
M1↑ | 3×Tg-AD | TNF-α GT | ↑ Microgliosis | ND | ND | ↓ | (Janelsins et al. 2008) |
M2a↑ | APP+PS1 | IL-4 GT | ↑ IL-4 ↓ Astro/microgliosis | ↑ | ↑ | ↓ | (Kiyota et al. 2010) |
M2c↑ | APP+PS1 | IL-10 GT | ↓ Astro/microgliosis | ↑ | ↑ | ↓ | (Kiyota et al. 2011) |
M2c↑ | APP | IL-10 GT | ↓ Microgliosis | ND | ↑ | ↓ | (Hara et al. 2011) |
M2c↑ | APP | × TGF-β1 TG | ↑ Microgliosis | ND | ND | ↓ | (Wyss-Coray et al. 2001) |
M2c↓ | APP | DN TGF-βR TG | ↑ Microgliosis | ND | ND | ↑ | (Tesseur et al. 2006) |
M2c↓ | APP | DN TGF-βR TG | ↑ Dendritic cells ↓ Astrogliosis | ND | ↑ | ↓ | (Town et al. 2008)) |
DN = Dominant negative, GT= Gene transfer, ND = Not determined, TG = Transgenic, ×= cross-bred
7. Neurogenesis
Adult neurogenesis is the process of neuronal proliferation and maturation in specific brain regions, and it is not only involved in CNS maintenance but also in the formation of new memories that new evidence suggests may be linked to synaptic plasticity affected by environmental or behavioral factors (Bruel-Jungerman et al., 2007; Deng et al., 2009; Deng et al., 2010; Tronel et al., 2010). Neural stem cells (NSCs) are always present in the adult CNS by self-renewal where they have the ability to differentiate into neurons, astrocytes, and oligodendrocytes. However, the process is strictly controlled by local signaling that keeps the production of new neurons to a very low level (Dayer et al., 2005; Tamura et al., 2007). The production of new neurons in adulthood is mainly seen in the subventricular zone (SVZ), where neuroblasts migrate to the olfactory bulb, and the subgranular zone (SGZ) of the hippocampal dentate gyrus, where the neurons are involved in hippocampal cell maintenance, the process of short-term memory formation and learning, and the regulation of mood (Ming and Song, 2005; Franklin and Ffrench-Constant, 2008). These areas have a network with high demand for the production of new neurons because they are constantly exposed to novel stimuli (Carpentier and Palmer, 2009). Only a small subset of NPCs created in the SGZ reach maturity after integration into the neural network, meaning a majority of the newborn cells undergo apoptosis within the first four days after creation (Sierra et al., 2010). It was shown by a number of researchers that exposure to novel smells in the olfactory bulb stimulated the production and retention of new neurons (Magavi et al., 2005; So et al., 2008), and it was likewise shown in the hippocampus that environmental enrichment, physical activity, and the act of learning promoted the production of new neurons (Emsley et al., 2005; Duan et al., 2008). Oddly enough, both depression and olfactory deficits are early indications of degeneration in both AD and Parkinson’s disease (PD) (Bruck et al., 2004; Hawkes, 2006; Gabryelewicz et al., 2007; Matsuda, 2007; Poewe, 2008). In fact, in co-cultures of resting or IL-4-activated microglia, NPCs expressing mutant PS1 variants have inhibited neuronal proliferation and differentiation compared to NPCs expressing wild-type human PS1, suggesting that there is a specific link between components causing AD and neurogenesis (Choi et al., 2008).
8. The Effect of Neuroinflammation on Neurogenesis
8.1 M1 microglial activation and neurogenesis
Neurogenesis can be challenged by a number of factors that induce the immune system response including chronic stress, depression, injury, and neuroinflammation. The chronic infusion or even single injection of M1 pro-inflammatory response-inducing ligand, such as LPS, has been shown to cause M1 microglial activation and acute neuroinflammation that caused the subsequent inhibition of neurogenesis in the hippocampi of adult rodents (Ekdahl et al., 2003; Monje et al., 2003; Walter et al., 2011). This M1-induced disruption of neurogenesis is very specific. Proliferation of NPCs is unaffected by neuroinflammation. As previously stated, a majority of newborn cells in the SGZ will undergo apoptosis within the first four days of life, and those apoptotic cells will be cleared by microglia through phagocytosis, a process unperturbed by inflammation (Sierra et al., 2010). For example, LPS-induced suppression of neurogenesis did not affect the production and proliferation of NSCs; it negatively affected NSC capability for maturation and survival (Ekdahl et al., 2003; Bastos et al., 2008), again suggesting that the pro-inflammatory cascade affects the maturation and survival of new neurons rather than their proliferation (Ekdahl et al., 2003; Monje et al., 2003; Bastos et al., 2008). IFN-γ gene expression in in vitro NSC cultures showed the same reduction in differentiation (Ekdahl et al., 2003; Monje et al., 2003; Walter et al., 2011). This disruption in survival may be due to the abnormal synaptic morphology of neurons differentiated during the inflammatory state (Jakubs et al., 2008). Another possibility is the mitochondrial damage caused by neuroinflammation. Neural progenitor cells (NPCs) require energy in the form of ATP produced by mitochondria to be able to differentiate into neurons (Bernstein and Bamburg, 2003). Pro-inflammatory molecules like TNF-α and IL-6 have been shown to contribute to mitochondrial dysfunction during brain inflammation both in vitro and in vivo (Stadler et al., 1992; Zell et al., 1997; Behrens et al., 2008; Samavati et al., 2008). This lack of proper mitochondrial production of ATP due to damage-induced up-regulation of pro-inflammatory factors may contribute to the decrease in neural differentiation in AD. The accumulation of toxic Aβ42 may also contribute to the reduction of NSC renewal in later inflammatory stages. In in vitro cultures of NPCs, it was found that aggregated Aβ42 inhibited neuronal differentiation (Haughey et al., 2002). Together, these studies suggest that, while AD pathology may at first enhance synthesis of new neurons as a compensatory mechanism to replace lost neurons, it may in turn reduce neuronal maturation.
8.2 M2 Microglial Activation and Neurogenesis
The suppression of M1 activation by NSAIDs, glatiramer acetate, or IL-4 gene delivery in the APP, PS1, or APP+PS1 mouse brain restored the suppression of neurogenesis seen in the SGZ (Wen et al., 2004; Chevallier et al., 2005; Butovsky et al., 2006a; Butovsky et al., 2006b; Kiyota et al., 2010). The M2 cytokines mediate the neuroprotective responses of microglia: IL-4 stimulation of microglia reduced the production of TNF-α and up-regulated production of IGF1, an important pro-neurogenic factor that induce subsequent neurogenesis (Butovsky et al., 2005; Butovsky et al., 2006b). In accord, we recently identified that IL-10-stimulated microglia enhanced proliferation but not differentiation of NSCs in vitro (Kiyota et al., 2011). As previously stated, IL-10 is an M2c ligand. Stimulation of microglia by an M2a ligand, IL-4, on the other hand, enhanced neural differentiation (Kiyota et al., 2010). Thus, combined treatment of these anti-inflammatory molecules with growth factors like IGF-1 or fibroblast growth factor-2 (FGF-2) may be able to restore neurogenesis, in terms of both proliferation and differentiation, and restore neurocognitive functionality in AD patients.
8.3 Neurogenesis in Aging and AD
One study recently demonstrated that an age-dependent increase in specific blood-borne factors associated with M1 microglia, as shown by the presence of the M1 marker, CCL11, may contribute to the inhibition of neurogenesis in healthy aging humans, as was seen when young mice were joined to the systemic environments of old mice by heterochronic parabiosis. This also resulted in decreased synaptic plasticity and impaired contextual fear conditioning, spatial memory, and learning (Villeda et al., 2011). There have been several contradictory reports that show how neurogenesis is particularly altered in the AD brain as well (for review, see (Waldau and Shetty, 2008)). Investigators have demonstrated that, while there is some reduction in neuronal maturation in the pre-senile post-mortem AD hippocampus, there is an increase in cellular proliferation in the hippocampus (Jin et al., 2004; Boekhoorn et al., 2006). Therefore, despite the supposed increase in neurogenesis due to increased proliferation, there is an overall reduction in neurogenesis because these newly-proliferated cells do not mature. The chronic neuroinflammation seen in AD has been shown to be the major contributor to overall suppression of neurogenesis in the SVZ and SGZ (Monje et al., 2003; Hoehn et al., 2005; Deng et al., 2010). The data from studies of microglial and inflammatory activation yielded complementary results to those found in mouse models above.
9. Neuron-Microglia Interaction through CD200 - CD200R Signaling
9.1 CD200 and CD200R
CD200 is a type I transmembrane glycoprotein with an immunoglobulin superfamily (IgSF) domain (Clark et al., 1985). It is expressed on a number of cells involved in the immune response, including T cells, B cells, and particularly in neurons in the brain (McMaster and Williams, 1979; Barclay, 1981; Webb and Barclay, 1984), and it is over-expressed in some forms of cancer cells as a means to avoid immune attack (Kretz-Rommel et al., 2007; Moreaux et al., 2008; Siva et al., 2008). Its short cytoplasmic tail suggests that its recognition domain is largely extracellular (Clark et al., 1985). The CD200R is expressed largely on cells of myeloid lineage, namely, microglia (Barclay, 1981; Koning et al., 2010). Like CD200, it is a cell surface glycoprotein with IgSF domains and a single transmembrane region (Wright et al., 2000). Unlike CD200, however, it has a large cytoplasmic region allowing for intracellular signaling initiated by tyrosine phosphorylation (Wright et al. 2000).
In order for CD200 to activate anti-inflammatory signals via CD200R, there must be an interaction between the extracellular domains of CD200 and CD200R (Hatherley and Barclay, 2004; Chen and Gorczynski, 2005). While the CD200R does not contain an immunotyrosine-based inhibitory motif (ITIM) in its cytoplasmic domain, it behaves as an inhibitory receptor on myeloid cells (Ravetch and Lanier, 2000). ITIMs are induced by ligands and lead to subsequent tyrosine phosphorylation, usually by Src family kinases, which allows for the recruitment of phosphatases with a Src homology 2 (SH2) domain (Ravetch and Lanier, 2000). CD200 interacts with CD200R through an immunoregulatory signal on its N-terminal Ig domain where tyrosine phosphorylation of the cytoplasmic NPxY motif on CD200R leads to a sequence of signaling events and interaction with PTB-binding domains with recruited adaptor proteins downstream of tyrosine kinase 1 and 2 (Dok1 and Dok2) (Wright et al., 2000; Hatherley and Barclay, 2004). After binding to Dok1 and Dok2, the SH2-containing inhibitory signaling molecule inositol 5-phosphatase (SHIP) and RAS p21 protein activator 1 (RasGAP) are recruited (Zhang et al., 2004). The CD200-CD200R interaction negatively modulates myeloid cells, supposedly through binding of Dok2 to phosphorylated NPxY and recruitment of RasGAP and subsequent inhibition of Erk, p38 MAPK, and JNK pathways (Mihrshahi et al., 2009). This is the signaling pathway by which CD200 activates its anti-inflammatory actions.
9.2 The Role of CD200 in Microglial Activation and AD
As stated above, CD200 is mainly expressed in neurons in the brain (Webb and Barclay, 1984), and the CD200R is expressed on astrocytes and cells of the myeloid lineage, including dendrites, macrophages, neurophils, and microglia (Hoek et al., 2000; Wright et al., 2000; Broderick et al., 2002; Wright et al., 2003). Both have wide distribution and expression throughout the brain. CD200 expression decreases with age where it is simultaneously associated with increased microglial activation (Lyons et al., 2007b). It is likewise decreased in the brains of AD patients and in the brains of Aβ-treated mice (Lyons et al., 2009; Walker et al., 2009). Interestingly, hippocampal slices from mice lacking CD200 also showed significant impairments in synaptic plasticity, specifically in LTP, tying the age-dependent decrease in CD200 with the decreased ability to form new memories in AD patients (Costello et al., 2011).
Several pieces of evidence point to the importance of CD200 in both the immune response and inflammation in AD. It has been demonstrated by many from both in vitro and in vivo models that CD200 is an important regulator of microglial activation and inflammation. Much of this research is generated from work on mouse models of inflammatory diseases, like the experimental autoimmune encephalomyelitis (EAE) model for MS or experimental autoimmune uveoretinitis (EAU) mice. Several studies have demonstrated in these mouse models that CD200 regulates CNS autoimmune disorders by showing that blockage of CD200 or CD200R causes increased inflammation and tissue damage while treatment with agonistic anti-CD200R antibodies ameliorated such symptoms (Banerjee and Dick, 2004; Chitnis et al., 2007; Copland et al., 2007). Other in vivo and in vitro data come from mice lacking the CD200 or CD200R gene (CD200−/− and CD200R−/−). In vitro data from blocked CD200R on macrophages expressing CD200 from EAU mice, for example, showed an enhancement of IFN-γ-induced release of IL-6 and neuronal cell death (Copland et al., 2007). Mice that were deficient in CD200 showed enhanced spontaneous inflammation and exaggerated inflammatory responses to injurious stimuli, like what one would observe in EAE (Hoek et al., 2000). Complimentary evidence showed that mice lacking CD200R expression had enhanced production of TNF-α and inflammatory responses to LPS, despite their continued expression of CD200 (Boudakov et al., 2007).
In addition to those findings from models of inflammation, other data show that there is a clear correlation between CD200 levels and other markers of pro- or anti-inflammation, namely IL-4. IL-4, the important anti-inflammatory signaling molecule for M2a activation of microglia, regulates the expression of downstream CD200. In both in vitro and in vivo studies from mouse and human tissue, it has been shown that a lack of IL-4 is correlated with a decrease in CD200 expression, and an increase in IL-4 results in an increase in CD200 expression (Lyons et al., 2007b; Lyons et al., 2009; Koning et al., 2010). In animal models of AD, where the age-related increase in microglial activation was accompanied by an age-related or Aβ-induced decrease in CD200 expression, IL-4 treatment was able to restore that deficit (Lyons et al., 2007b; Lyons et al., 2007a; Kiyota et al., 2010). These tissues and glia from AD models lack IL-4 and have enhanced inflammatory responses to LPS (Lyons et al., 2009). As mentioned earlier, IL-4 gene therapy to APP+PS1 mice enhanced clearance of Aβ and improved spatial learning, but only in the pre-symptomatic stages of the disease (Kiyota et al., 2010). This suggests that IL-4 signaling may already be compromised at the later disease stages due to the decreased expression of CD200 in the aged and the AD brain. This is the reason why CD200 is a more plausible therapeutic target than IL-4, and may have a therapeutic effect on post-symptomatic AD mouse models.
Worth mentioning are findings from other disease models that allude to the importance of CD200. A number of different cancer cells over-express CD200, theoretically as a means to alleviate T-cell-mediated cytotoxicity (Kretz-Rommel et al., 2007; Moreaux et al., 2008; Siva et al., 2008). Melanoma cells had increased CD200 expression, and this was correlated with their potential to be metastatic. It was shown that cancer patients’ myeloma CD200 expression in turn correlated with their likelihood for survival (Moreaux et al., 2006; Petermann et al., 2007; Siva et al., 2008). This makes CD200 an excellent target in cancer therapy as well. It may be possible to target cancer cells over-expressing CD200 with an anti-CD200 antibody. These and all of the above findings show that there are good reasons for further research to investigate the role of CD200 in the brain. While there are some clear correlations seen in aging and in disease models, such as MS, AD, and cancer, the connection between CD200 and these diseases is still unclear. CD200 has never been tested for its therapeutic potential in AD animal models. The other issue brought to light by the results of the afore-mentioned IL-4 study is the relationship between neuroinflammation and neurogenesis in AD. Our lab and other researchers are currently looking into potential CD200 drug treatments or animal models to figure out the connection between IL-4 signaling, CD200, and neurogenesis in AD.
9.3 The Role of Dok in CD200R Signaling
One possible mechanism to look into is the Dok signaling pathway of the CD200R. As previously stated, when CD200 binds to CD200R, the interaction negatively modulates myeloid cells through binding of Dok2. While there has not been very much research on Dok proteins, they are involved in immune suppression signaling. They are expressed on peripheral T cells and most thymocytes, and act as negative regulators of T cell response signaling (Yasuda et al., 2007). Mice that lack Dok1 and/or Dok2 showed increased production of pro-inflammatory molecules, like TNF-α and NO, and enhanced responses to inflammation-inducing signals like those from IFN-γ and LPS (Shinohara et al., 2005; Yasuda et al., 2007).
These findings are similar to those seen in CD200−/− mice whose lack of ligand or receptor expression correlated with increased neuroinflammation and tissue damage and production of TNF-α in response to LPS. There is no published study to date that shows the regulating relationship between CD200R and Dok proteins in microglia and inflammation. CD200R is believed to act via Dok2 signaling, but it is still unknown which is the controlling mechanism. Figure 1 outlines the unresolved pathways that CD200/CD200R/Dok2 signaling may follow to attain its anti-inflammatory effects. While Dok1/2−/− mice seem to have similar phenotypes to CD200 or CD200R−/− mice, it has not yet been tested whether treatment of Dok1/2−/− mice with CD200 can compensate for maintenance of immune system homeostasis (Fig. 1).
One important issue worthy of mentioning, however, is the limitations of using mouse models for modeling human diseases. Koning et al did a comparative study of CD200R expression in the mouse and the human, and found that, while induction of CD200R expression in human macrophages was dependent on IL-4, mouse CD200R expression was not (Koning et al., 2010). Induction of mouse CD200R expression was not discovered, but it was theorized that it involved site-specific combinations of cytokines (Snelgrove et al., 2008). Therefore, while the CD200R may be a suitable indicator of alternative activation in humans, this may be a species-specific mechanism. This shows that research into the controlling mechanism between CD200R and Dok signaling may be a key finding.
10. Concluding Remarks
Research has still been unable to pinpoint a single cause for the neuronal death and synaptic dysfunction that leads to the cognitive decline seen in AD. Increasing evidence suggests that the cause is an interaction between the down-regulation of important immune regulatory components, increased inflammation, and decreased neurogenesis, and it is becoming more widely believed that inflammation is the major culprit in this interaction. This neuroinflammation is orchestrated by M1-skewed microglial activation often associated with loss of neurons in specific brain regions due to the production of damaging reactive oxygen species and toxic intermediates (Gutteridge and Halliwell 1989; Pratico et al. 2001; Pratico and Sung 2004). Alternative M2 microglial activation, on the other hand, is important for resolution of this inflammation. M2 activation causes the release of anti-inflammatory cytokines like IL-4, which is important for M2a microglial activation, and IL-10, which is important for M2c.
There is a characteristic switch from M2 to M1 microglial activation in the brain that occurs both with age and with disease progression. There is an up-regulation of pro-inflammatory cytokines and a reduction in anti-inflammatory cytokines and proteins, like CD200 (Lyons et al., 2007b; Lyons et al., 2009; Walker et al., 2009). CD200 interacts with CD200R via the Dok signaling pathway to activate anti-inflammatory signals, and its age-dependent decline is associated with increased microglial activation (Lyons et al., 2007b). A lack of CD200 or Dok results in increased neuroinflammation in the brain (Shinohara et al., 2005; Yasuda et al., 2007). This is seen in both rodent and human brain tissue. Treatments with specific M2a or M2c anti-inflammatory cytokines, like IL-4 or IL-10, can improve deficits seen as a result of up-regulated inflammatory responses, and restore CD200 expression which is specific to IL-4 (Lyons et al., 2007b; Lyons et al., 2007a; Kiyota et al., 2010). Neuroinflammatory states have also been shown to greatly decrease neurogenesis in the SVZ and SGZ, which may explain the deficit in short-term memory formation in AD patients. Evidence suggests that M1 microglial activation suppresses, not the proliferation of NSCs, but the differentiation of NPCs (Waldau and Shetty, 2008). Treatment with M2 cytokines or proteins like CD200, IL-4, or IL-10, and growth factors like FGF-2 can possibly restore that deficit.
There is still no successful therapy available to halt and reverse the devastating neurodegeneration seen in AD, but the evidence leads one to conclude that the best treatment for AD may be a combined therapeutic with anti-inflammatory and pro-neurogenic components. While none have been thoroughly tested, IL-4, IL-10, CD200, FGF-2, and Dok signaling proteins have proven to be promising targets for further research. Regardless of the various potentials of these therapeutic targets, it is clear that there is still a lack of understanding of the complicated relationship between inflammation and AD that warrants further investigation.
Acknowledgement
We would like to thank Seiko Ikezu and Maya Woodbury for editing the manuscript. This work was supported in part by National Institute of Health grants 5T32GM008541 to MMV, and MH072539 and AG032600 to TI.
Abbreviations
Aβ | amyloid β-peptide |
AD | Alzheimer’s disease |
ADAM | a disintegrin and metalloproteinase |
AICD | APP intracellular domain |
APP | amyloid precursor protein |
ARG1 | arginase 1 |
BACE | β-site APP converting enzyme |
BrdU | bromodeoxyuridine |
CD200 | cluster of differentiation 200 (aka OX2) |
CNS | central nervous system |
DAMP | damage-associated molecular pattern |
Dok | downstream of tyrosine kinase |
Erk | extracellular signal-regulated kinase |
EAE | experimental autoimmune encephalomyelitis |
EAU | experimental autoimmune uveoretinitis |
FasL | Fas ligand |
FGF-2 | fibroblast growth factor-2 |
FIZZ1 | found in inflammatory zone 1 |
HPA | hypothalamo-pituitary-adrenal |
IGF-1 | insulin-like growth factor-1 |
IgSF | immunoglobulin superfamily |
IFN-γ | interferon-γ |
IL | interleukin |
ITIM | immunotyrosine-based inhibitory motif |
JNK | c-Jun N-terminal kinase |
LPS | lipopolysaccharide |
LTP | long-term potentiation |
MAPK | mitogen-activated protein kinase |
MDP | muramyl dipeptide |
MR | mannose receptor |
MS | multiple sclerosis |
NFT | neurofibrillary tangle |
NLR | NOD-like receptor |
NO | nitric oxide |
NOD | nucleotide oligomerization domain |
NOS | nitric oxide synthase |
NPC | neural progenitor cell |
NSAID | non-steroidal anti-inflammatory drug |
NSC | neural stem cell |
PAMP | pathogen-associated molecular pattern |
PD | Parkinson’s disease |
PEN-2 | presenilin enhancer-2 |
PRR | pathogen recognition receptor |
PS | presenilin |
RasGAP RAS | p21 protein activator 1 |
RIG1 | retinoic acid-inducible gene-1 |
RLR | RIG1-like receptor |
SGZ | subgranular zone |
SH2 | Src homology 2 |
SHIP | SH2-containing inhibitory signaling molecule inositol 5-phosphatase |
SVZ | subventricular zone |
TGF-β | transforming growth factor-β |
TLR | toll-like receptor |
TNF-α | tumor necrosis factor-α |
YM1 | chitinase 3-like 3 |
WT | wild type |
References
- Abbas N, Bednar I, Mix E, Marie S, Paterson D, Ljungberg A, Morris C, Winblad B, Nordberg A, Zhu J. Up-regulation of the inflammatory cytokines IFN-gamma and IL-12 and down-regulation of IL-4 in cerebral cortex regions of APP(SWE) transgenic mice. J Neuroimmunol. 2002;126:50–57. [Abstract] [Google Scholar]
- Aizenstein HJ, Nebes RD, Saxton JA, Price JC, Mathis CA, Tsopelas ND, Ziolko SK, James JA, Snitz BE, Houck PR, Bi W, Cohen AD, Lopresti BJ, DeKosky ST, Halligan EM, Klunk WE. Frequent amyloid deposition without significant cognitive impairment among the elderly. Archives of neurology. 2008;65:1509–1517. [Europe PMC free article] [Abstract] [Google Scholar]
- Akiyama H, Barger S, Barnum S, Bradt B, Bauer J, Cole GM, Cooper NR, Eikelenboom P, Emmerling M, Fiebich BL, Finch CE, Frautschy S, Griffin WS, Hampel H, Hull M, Landreth G, Lue L, Mrak R, Mackenzie IR, McGeer PL, O’Banion MK, Pachter J, Pasinetti G, Plata-Salaman C, Rogers J, Rydel R, Shen Y, Streit W, Strohmeyer R, Tooyoma I, Van Muiswinkel FL, Veerhuis R, Walker D, Webster S, Wegrzyniak B, Wenk G, Wyss-Coray T. Inflammation and Alzheimer’s disease. Neurobiol Aging. 2000;21:383–421. [Europe PMC free article] [Abstract] [Google Scholar]
- Alzheimer A, Stelzmann RA, Schnitzlein HN, Murtagh FR. An English translation of Alzheimer’s 1907 paper, “Uber eine eigenartige Erkankung der Hirnrinde” Clin Anat. 1995;8:429–431. [Abstract] [Google Scholar]
- Apelt J, Schliebs R. Beta-amyloid-induced glial expression of both pro- and anti-inflammatory cytokines in cerebral cortex of aged transgenic Tg2576 mice with Alzheimer plaque pathology. Brain Res. 2001;894:21–30. [Abstract] [Google Scholar]
- Baek SH, Ohgi KA, Rose DW, Koo EH, Glass CK, Rosenfeld MG. Exchange of N-CoR corepressor and Tip60 coactivator complexes links gene expression by NF-kappaB and beta-amyloid precursor protein. Cell. 2002;110:55–67. [Abstract] [Google Scholar]
- Bailey SL, Carpentier PA, McMahon EJ, Begolka WS, Miller SD. Innate and adaptive immune responses of the central nervous system. Crit Rev Immunol. 2006;26:149–188. [Abstract] [Google Scholar]
- Baker BJ, Akhtar LN, Benveniste EN. SOCS1 and SOCS3 in the control of CNS immunity. Trends Immunol. 2009;30:392–400. [Europe PMC free article] [Abstract] [Google Scholar]
- Banerjee D, Dick AD. Blocking CD200-CD200 receptor axis augments NOS-2 expression and aggravates experimental autoimmune uveoretinitis in Lewis rats. Ocul Immunol Inflamm. 2004;12:115–125. [Abstract] [Google Scholar]
- Barclay AN. Different reticular elements in rat lymphoid tissue identified by localization of Ia, Thy-1 and MRC OX 2 antigens. Immunology. 1981;44:727–736. [Abstract] [Google Scholar]
- Bastos GN, Moriya T, Inui F, Katura T, Nakahata N. Involvement of cyclooxygenase-2 in lipopolysaccharide-induced impairment of the newborn cell survival in the adult mouse dentate gyrus. Neuroscience. 2008;155:454–462. [Abstract] [Google Scholar]
- Bauer J, Strauss S, Schreiter-Gasser U, Ganter U, Schlegel P, Witt I, Yolk B, Berger M. Interleukin-6 and alpha-2-macroglobulin indicate an acute-phase state in Alzheimer’s disease cortices. FEBS Lett. 1991;285:111–114. [Abstract] [Google Scholar]
- Bauer J, Ganter U, Strauss S, Stadtmuller G, Frommberger U, Bauer H, Volk B, Berger M. The participation of interleukin-6 in the pathogenesis of Alzheimer’s disease. Res Immunol. 1992;143:650–657. [Abstract] [Google Scholar]
- Behrens MM, Ali SS, Dugan LL. Interleukin-6 mediates the increase in NADPH-oxidase in the ketamine model of schizophrenia. J Neurosci. 2008;28:13957–13966. [Europe PMC free article] [Abstract] [Google Scholar]
- Bennett DA, Schneider JA, Arvanitakis Z, Kelly JF, Aggarwal NT, Shah RC, Wilson RS. Neuropathology of older persons without cognitive impairment from two community-based studies. Neurology. 2006;66:1837–1844. [Abstract] [Google Scholar]
- Benoit M, Desnues B, Mege JL. Macrophage polarization in bacterial infections. J Immunol. 2008;181:3733–3739. [Abstract] [Google Scholar]
- Benzing WC, Wujek JR, Ward EK, Shaffer D, Ashe KH, Younkin SG, Brunden KR. Evidence for glial-mediated inflammation in aged APP(SW) transgenic mice. Neurobiol Aging. 1999;20:581–589. [Abstract] [Google Scholar]
- Bernstein BW, Bamburg JR. Actin-ATP hydrolysis is a major energy drain for neurons. J Neurosci. 2003;23:1–6. [Europe PMC free article] [Abstract] [Google Scholar]
- Bird TD. Genetic aspects of Alzheimer disease. Genet Med. 2008;10:231–239. [Europe PMC free article] [Abstract] [Google Scholar]
- Bishop NA, Lu T, Yankner BA. Neural mechanisms of ageing and cognitive decline. Nature. 2010;464:529–535. [Europe PMC free article] [Abstract] [Google Scholar]
- Boekhoorn K, Joels M, Lucassen PJ. Increased proliferation reflects glial and vascular-associated changes, but not neurogenesis in the presenile Alzheimer hippocampus. Neurobiology of disease. 2006;24:1–14. [Abstract] [Google Scholar]
- Boudakov I, Liu J, Fan N, Gulay P, Wong K, Gorczynski RM. Mice lacking CD200R1 show absence of suppression of lipopolysaccharide-induced tumor necrosis factor-alpha and mixed leukocyte culture responses by CD200. Transplantation. 2007;84:251–257. [Abstract] [Google Scholar]
- Broderick C, Hoek RM, Forrester JV, Liversidge J, Sedgwick JD, Dick AD. Constitutive retinal CD200 expression regulates resident microglia and activation state of inflammatory cells during experimental autoimmune uveoretinitis. Am J Pathol. 2002;161:1669–1677. [Europe PMC free article] [Abstract] [Google Scholar]
- Brown GC. Mechanisms of inflammatory neurodegeneration: iNOS and NADPH oxidase. Biochem Soc Trans. 2007;35:1119–1121. [Abstract] [Google Scholar]
- Bruck A, Kurki T, Kaasinen V, Vahlberg T, Rinne JO. Hippocampal and prefrontal atrophy in patients with early non-demented Parkinson’s disease is related to cognitive impairment. J Neurol Neurosurg Psychiatry. 2004;75:1467–1469. [Europe PMC free article] [Abstract] [Google Scholar]
- Bruel-Jungerman E, Rampon C, Laroche S. Adult hippocampal neurogenesis, synaptic plasticity and memory: facts and hypotheses. Rev Neurosci. 2007;18:93–114. [Abstract] [Google Scholar]
- Bsibsi M, Ravid R, Gveric D, van Noort JM. Broad expression of Toll-like receptors in the human central nervous system. J Neuropathol Exp Neurol. 2002;61:1013–1021. [Abstract] [Google Scholar]
- Burton MD, Sparkman NL, Johnson RW. Inhibition of interleukin-6 trans-signaling in the brain facilitates recovery from lipopolysaccharide-induced sickness behavior. J Neuroinflammation. 2011;8:54. [Europe PMC free article] [Abstract] [Google Scholar]
- Butovsky O, Talpalar AE, Ben-Yaakov K, Schwartz M. Activation of microglia by aggregated beta-amyloid or lipopolysaccharide impairs MHC-II expression and renders them cytotoxic whereas IFN-gamma and IL-4 render them protective. Mol Cell Neurosci. 2005;29:381–393. [Abstract] [Google Scholar]
- Butovsky O, Koronyo-Hamaoui M, Kunis G, Ophir E, Landa G, Cohen H, Schwartz M. Glatiramer acetate fights against Alzheimer’s disease by inducing dendritic-like microglia expressing insulin-like growth factor 1. Proc Natl Acad Sci U S A. 2006a;103:11784–11789. [Europe PMC free article] [Abstract] [Google Scholar]
- Butovsky O, Ziv Y, Schwartz A, Landa G, Talpalar AE, Pluchino S, Martino G, Schwartz M. Microglia activated by IL-4 or IFN-gamma differentially induce neurogenesis and oligodendrogenesis from adult stem/progenitor cells. Mol Cell Neurosci. 2006b;31:149–160. [Abstract] [Google Scholar]
- Cao X, Sudhof TC. A transcriptionally [correction of transcriptively] active complex of APP with Fe65 and histone acetyltransferase Tip60. Science. 2001;293:115–120. [Abstract] [Google Scholar]
- Cao X, Sudhof TC. Dissection of amyloid-beta precursor protein-dependent transcriptional transactivation. J Biol Chem. 2004;279:24601–24611. [Abstract] [Google Scholar]
- Carpentier PA, Palmer TD. Immune influence on adult neural stem cell regulation and function. Neuron. 2009;64:79–92. [Europe PMC free article] [Abstract] [Google Scholar]
- Chandler S, Coates R, Gearing A, Lury J, Wells G, Bone E. Matrix metalloproteinases degrade myelin basic protein. Neurosci Lett. 1995;201:223–226. [Abstract] [Google Scholar]
- Chauhan VS, Sterka DG, Jr., Furr SR, Young AB, Marriott I. NOD2 plays an important role in the inflammatory responses of microglia and astrocytes to bacterial CNS pathogens. Glia. 2009;57:414–423. [Europe PMC free article] [Abstract] [Google Scholar]
- Chen DX, Gorczynski RM. Discrete monoclonal antibodies define functionally important epitopes in the CD200 molecule responsible for immunosuppression function. Transplantation. 2005;79:282–288. [Abstract] [Google Scholar]
- Chevallier NL, Soriano S, Kang DE, Masliah E, Hu G, Koo EH. Perturbed neurogenesis in the adult hippocampus associated with presenilin-1 A246E mutation. Am J Pathol. 2005;167:151–159. [Europe PMC free article] [Abstract] [Google Scholar]
- Chitnis T, Imitola J, Wang Y, Elyaman W, Chawla P, Sharuk M, Raddassi K, Bronson RT, Khoury SJ. Elevated neuronal expression of CD200 protects Wlds mice from inflammation-mediated neurodegeneration. Am J Pathol. 2007;170:1695–1712. [Europe PMC free article] [Abstract] [Google Scholar]
- Choi SH, Veeraraghavalu K, Lazarov O, Marler S, Ransohoff RM, Ramirez JM, Sisodia SS. Non-cell-autonomous effects of presenilin 1 variants on enrichment-mediated hippocampal progenitor cell proliferation and differentiation. Neuron. 2008;59:568–580. [Europe PMC free article] [Abstract] [Google Scholar]
- Citron M, Oltersdorf T, Haass C, McConlogue L, Hung AY, Seubert P, Vigo-Pelfrey C, Lieberburg I, Selkoe DJ. Mutation of the beta-amyloid precursor protein in familial Alzheimer’s disease increases beta-protein production. Nature. 1992;360:672–674. [Abstract] [Google Scholar]
- Clark MJ, Gagnon J, Williams AF, Barclay AN. MRC OX-2 antigen: a lymphoid/neuronal membrane glycoprotein with a structure like a single immunoglobulin light chain. EMBO J. 1985;4:113–118. [Europe PMC free article] [Abstract] [Google Scholar]
- Cole GM, Frautschy SA. Mechanisms of action of non-steroidal anti-inflammatory drugs for the prevention of Alzheimer’s disease. CNS Neurol Disord Drug Targets. 2010;9:140–148. [Europe PMC free article] [Abstract] [Google Scholar]
- Colton CA. Heterogeneity of microglial activation in the innate immune response in the brain. J Neuroimmune Pharmacol. 2009;4:399–418. [Europe PMC free article] [Abstract] [Google Scholar]
- Colton CA, Mott RT, Sharpe H, Xu Q, Van Nostrand WE, Vitek MP. Expression profiles for macrophage alternative activation genes in AD and in mouse models of AD. J Neuroinflammation. 2006a;3:27. [Europe PMC free article] [Abstract] [Google Scholar]
- Colton CA, Wilcock DM, Wink DA, Davis J, Van Nostrand WE, Vitek MP. The effects of NOS2 gene deletion on mice expressing mutated human AbetaPP. J Alzheimers Dis. 2008;15:571–587. [Europe PMC free article] [Abstract] [Google Scholar]
- Colton CA, Vitek MP, Wink DA, Xu Q, Cantillana V, Previti ML, Van Nostrand WE, Weinberg JB, Dawson H. NO synthase 2 (NOS2) deletion promotes multiple pathologies in a mouse model of Alzheimer’s disease. Proc Natl Acad Sci U S A. 2006b;103:12867–12872. [Europe PMC free article] [Abstract] [Google Scholar]
- Copland DA, Calder CJ, Raveney BJ, Nicholson LB, Phillips J, Cherwinski H, Jenmalm M, Sedgwick JD, Dick AD. Monoclonal antibody-mediated CD200 receptor signaling suppresses macrophage activation and tissue damage in experimental autoimmune uveoretinitis. Am J Pathol. 2007;171:580–588. [Europe PMC free article] [Abstract] [Google Scholar]
- Costello DA, Lyons A, Browne T, Denieffe S, Cox FF, Lynch MA. Long-term potentiation is impaired in CD200-deficient mice: a role for Toll-like receptor activation. J Biol Chem. 2011 [Europe PMC free article] [Abstract] [Google Scholar]
- David JR. Macrophage activation induced by lymphocyte mediators. Acta Endocrinol Suppl (Copenh) 1975;194:245–261. [Abstract] [Google Scholar]
- Davies CA, Mann DM, Sumpter PQ, Yates PO. A quantitative morphometric analysis of the neuronal and synaptic content of the frontal and temporal cortex in patients with Alzheimer’s disease. J Neurol Sci. 1987;78:151–164. [Abstract] [Google Scholar]
- Dayer AG, Cleaver KM, Abouantoun T, Cameron HA. New GABAergic interneurons in the adult neocortex and striatum are generated from different precursors. J Cell Biol. 2005;168:415–427. [Europe PMC free article] [Abstract] [Google Scholar]
- Deng W, Aimone JB, Gage FH. New neurons and new memories: how does adult hippocampal neurogenesis affect learning and memory? Nat Rev Neurosci. 2010;11:339–350. [Europe PMC free article] [Abstract] [Google Scholar]
- Deng W, Saxe MD, Gallina IS, Gage FH. Adult-born hippocampal dentate granule cells undergoing maturation modulate learning and memory in the brain. J Neurosci. 2009;29:13532–13542. [Europe PMC free article] [Abstract] [Google Scholar]
- Dickson DW, Farlo J, Davies P, Crystal H, Fuld P, Yen SH. Alzheimer’s disease. A double-labeling immunohistochemical study of senile plaques. Am J Pathol. 1988;132:86–101. [Europe PMC free article] [Abstract] [Google Scholar]
- Duan X, Kang E, Liu CY, Ming GL, Song H. Development of neural stem cell in the adult brain. Curr Opin Neurobiol. 2008;18:108–115. [Europe PMC free article] [Abstract] [Google Scholar]
- Edison P, Archer HA, Gerhard A, Hinz R, Pavese N, Turkheimer FE, Hammers A, Tai YF, Fox N, Kennedy A, Rossor M, Brooks DJ. Microglia, amyloid, and cognition in Alzheimer’s disease: An [11C](R)PK11195-PET and [11C]PIB-PET study. Neurobiology of disease. 2008;32:412–419. [Abstract] [Google Scholar]
- Ekdahl CT, Claasen JH, Bonde S, Kokaia Z, Lindvall O. Inflammation is detrimental for neurogenesis in adult brain. Proc Natl Acad Sci U S A. 2003;100:13632–13637. [Europe PMC free article] [Abstract] [Google Scholar]
- Elenkov IJ, Chrousos GP. Stress hormones, proinflammatory and antiinflammatory cytokines, and autoimmunity. Ann N Y Acad Sci. 2002;966:290–303. [Abstract] [Google Scholar]
- Emsley JG, Mitchell BD, Kempermann G, Macklis JD. Adult neurogenesis and repair of the adult CNS with neural progenitors, precursors, and stem cells. Prog Neurobiol. 2005;75:321–341. [Abstract] [Google Scholar]
- Esch FS, Keim PS, Beattie EC, Blacher RW, Culwell AR, Oltersdorf T, McClure D, Ward PJ. Cleavage of amyloid beta peptide during constitutive processing of its precursor. Science. 1990;248:1122–1124. [Abstract] [Google Scholar]
- Figueiredo C, Pais TF, Gomes JR, Chatterjee S. Neuron-microglia crosstalk up-regulates neuronal FGF-2 expression which mediates neuroprotection against excitotoxicity via JNK1/2. J Neurochem. 2008;107:73–85. [Abstract] [Google Scholar]
- Fillit H, Ding WH, Buee L, Kalman J, Altstiel L, Lawlor B, Wolf-Klein G. Elevated circulating tumor necrosis factor levels in Alzheimer’s disease. Neurosci Lett. 1991;129:318–320. [Abstract] [Google Scholar]
- Firuzi O, Pratico D. Coxibs and Alzheimer’s disease: should they stay or should they go? Ann Neurol. 2006;59:219–228. [Abstract] [Google Scholar]
- Franklin RJ, Ffrench-Constant C. Remyelination in the CNS: from biology to therapy. Nat Rev Neurosci. 2008;9:839–855. [Abstract] [Google Scholar]
- Gabryelewicz T, Styczynska M, Luczywek E, Barczak A, Pfeffer A, Androsiuk W, Chodakowska-Zebrowska M, Wasiak B, Peplonska B, Barcikowska M. The rate of conversion of mild cognitive impairment to dementia: predictive role of depression. Int J Geriatr Psychiatry. 2007;22:563–567. [Abstract] [Google Scholar]
- Gomi M, Aoki T, Takagi Y, Nishimura M, Ohsugi Y, Mihara M, Nozaki K, Hashimoto N, Miyamoto S, Takahashi J. Single and local blockade of interleukin-6 signaling promotes neuronal differentiation from transplanted embryonic stem cell-derived neural precursor cells. J Neurosci Res. 2011;89:1388–1399. [Abstract] [Google Scholar]
- Gordon S. Alternative activation of macrophages. Nat Rev Immunol. 2003;3:23–35. [Abstract] [Google Scholar]
- Gutowicz M. The influence of reactive oxygen species on the central nervous system. Postepy Hig Med Dosw (Online) 2011;65:104–113. [Abstract] [Google Scholar]
- Gutteridge JM, Halliwell B. Iron toxicity and oxygen radicals. Baillieres Clin Haematol. 1989;2:195–256. [Abstract] [Google Scholar]
- Haass C, De Strooper B. The presenilins in Alzheimer’s disease--proteolysis holds the key. Science. 1999;286:916–919. [Abstract] [Google Scholar]
- Haga S, Akai K, Ishii T. Demonstration of microglial cells in and around senile (neuritic) plaques in the Alzheimer brain. An immunohistochemical study using a novel monoclonal antibody. Acta Neuropathol. 1989;77:569–575. [Abstract] [Google Scholar]
- Haigis MC, Yankner BA. The aging stress response. Molecular cell. 2010;40:333–344. [Europe PMC free article] [Abstract] [Google Scholar]
- Hara H, Mouri A, Yonemitsu Y, Nabeshima T, Tabira T. Mucosal immunotherapy in an Alzheimer mouse model by recombinant Sendai virus vector carrying Abeta1-43/IL-10 cDNA. Vaccine. 2011;29:7474–7482. [Abstract] [Google Scholar]
- Hardy J, Selkoe DJ. The amyloid hypothesis of Alzheimer’s disease: progress and problems on the road to therapeutics. Science. 2002;297:353–356. [Abstract] [Google Scholar]
- Hatherley D, Barclay AN. The CD200 and CD200 receptor cell surface proteins interact through their N-terminal immunoglobulin-like domains. Eur J Immunol. 2004;34:1688–1694. [Abstract] [Google Scholar]
- Haughey NJ, Nath A, Chan SL, Borchard AC, Rao MS, Mattson MP. Disruption of neurogenesis by amyloid beta-peptide, and perturbed neural progenitor cell homeostasis, in models of Alzheimer’s disease. J Neurochem. 2002;83:1509–1524. [Abstract] [Google Scholar]
- Hawkes C. Olfaction in neurodegenerative disorder. Adv Otorhinolaryngol. 2006;63:133–151. [Abstract] [Google Scholar]
- He P, Zhong Z, Lindholm K, Berning L, Lee W, Lemere C, Staufenbiel M, Li R, Shen Y. Deletion of tumor necrosis factor death receptor inhibits amyloid beta generation and prevents learning and memory deficits in Alzheimer’s mice. J Cell Biol. 2007;178:829–841. [Europe PMC free article] [Abstract] [Google Scholar]
- Hein AM, Stasko MR, Matousek SB, Scott-McKean JJ, Maier SF, Olschowka JA, Costa AC, O’Banion MK. Sustained hippocampal IL-1beta overexpression impairs contextual and spatial memory in transgenic mice. Brain Behav Immun. 2010;24:243–253. [Europe PMC free article] [Abstract] [Google Scholar]
- Heinrich PC, Behrmann I, Muller-Newen G, Schaper F, Graeve L. Interleukin-6-type cytokine signalling through the gp130/Jak/STAT pathway. Biochem J. 1998;334(Pt 2):297–314. [Europe PMC free article] [Abstract] [Google Scholar]
- Hoehn BD, Palmer TD, Steinberg GK. Neurogenesis in rats after focal cerebral ischemia is enhanced by indomethacin. Stroke. 2005;36:2718–2724. [Abstract] [Google Scholar]
- Hoek RM, Ruuls SR, Murphy CA, Wright GJ, Goddard R, Zurawski SM, Blom B, Homola ME, Streit WJ, Brown MH, Barclay AN, Sedgwick JD. Down-regulation of the macrophage lineage through interaction with OX2 (CD200) Science. 2000;290:1768–1771. [Abstract] [Google Scholar]
- Holmes C, Boche D, Wilkinson D, Yadegarfar G, Hopkins V, Bayer A, Jones RW, Bullock R, Love S, Neal JW, Zotova E, Nicoll JA. Long-term effects of Abeta42 immunisation in Alzheimer’s disease: follow-up of a randomised, placebo-controlled phase I trial. Lancet. 2008;372:216–223. [Abstract] [Google Scholar]
- Hoozemans JJ, Veerhuis R, Rozemuller JM, Eikelenboom P. Neuroinflammation and regeneration in the early stages of Alzheimer’s disease pathology. Int J Dev Neurosci. 2006;24:157–165. [Abstract] [Google Scholar]
- Iqbal K, Alonso AC, Gong CX, Khatoon S, Pei JJ, Wang JZ, Grundke-Iqbal I. Mechanisms of neurofibrillary degeneration and the formation of neurofibrillary tangles. J Neural Transm Suppl. 1998;53:169–180. [Abstract] [Google Scholar]
- Ishii T, Haga S. Complements, microglial cells and amyloid fibril formation. Res Immunol. 1992;143:614–616. [Abstract] [Google Scholar]
- Iwatsubo T, Mann DM, Odaka A, Suzuki N, Ihara Y. Amyloid beta protein (A beta) deposition: A beta 42(43) precedes A beta 40 in Down syndrome. Ann Neurol. 1995;37:294–299. [Abstract] [Google Scholar]
- Jakubs K, Bonde S, Iosif RE, Ekdahl CT, Kokaia Z, Kokaia M, Lindvall O. Inflammation regulates functional integration of neurons born in adult brain. J Neurosci. 2008;28:12477–12488. [Europe PMC free article] [Abstract] [Google Scholar]
- Janelsins MC, Mastrangelo MA, Oddo S, LaFerla FM, Federoff HJ, Bowers WJ. Early correlation of microglial activation with enhanced tumor necrosis factor-alpha and monocyte chemoattractant protein-1 expression specifically within the entorhinal cortex of triple transgenic Alzheimer’s disease mice. J Neuroinflammation. 2005;2:23. [Europe PMC free article] [Abstract] [Google Scholar]
- Jankowsky JL, Fadale DJ, Anderson J, Xu GM, Gonzales V, Jenkins NA, Copeland NG, Lee MK, Younkin LH, Wagner SL, Younkin SG, Borchelt DR. Mutant presenilins specifically elevate the levels of the 42 residue beta-amyloid peptide in vivo: evidence for augmentation of a 42-specific gamma secretase. Hum Mol Genet. 2004;13:159–170. [Abstract] [Google Scholar]
- Jarrett JT, Berger EP, Lansbury PT., Jr. The carboxy terminus of the beta amyloid protein is critical for the seeding of amyloid formation: implications for the pathogenesis of Alzheimer’s disease. Biochemistry. 1993;32:4693–4697. [Abstract] [Google Scholar]
- Jimenez S, Baglietto-Vargas D, Caballero C, Moreno-Gonzalez I, Torres M, Sanchez-Varo R, Ruano D, Vizuete M, Gutierrez A, Vitorica J. Inflammatory response in the hippocampus of PS1M146L/APP751SL mouse model of Alzheimer’s disease: age-dependent switch in the microglial phenotype from alternative to classic. J Neurosci. 2008;28:11650–11661. [Europe PMC free article] [Abstract] [Google Scholar]
- Jin JJ, Kim HD, Maxwell JA, Li L, Fukuchi K. Toll-like receptor 4-dependent upregulation of cytokines in a transgenic mouse model of Alzheimer’s disease. J Neuroinflammation. 2008;5:23. [Europe PMC free article] [Abstract] [Google Scholar]
- Jin K, Peel AL, Mao XO, Xie L, Cottrell BA, Henshall DC, Greenberg DA. Increased hippocampal neurogenesis in Alzheimer’s disease. Proc Natl Acad Sci U S A. 2004;101:343–347. [Europe PMC free article] [Abstract] [Google Scholar]
- Kadir A, Marutle A, Gonzalez D, Scholl M, Almkvist O, Mousavi M, Mustafiz T, Darreh-Shori T, Nennesmo I, Nordberg A. Positron emission tomography imaging and clinical progression in relation to molecular pathology in the first Pittsburgh Compound B positron emission tomography patient with Alzheimer’s disease. Brain: a journal of neurology. 2011;134:301–317. [Europe PMC free article] [Abstract] [Google Scholar]
- Kimberly WT, LaVoie MJ, Ostaszewski BL, Ye W, Wolfe MS, Selkoe DJ. Gamma-secretase is a membrane protein complex comprised of presenilin, nicastrin, Aph-1, and Pen-2. Proc Natl Acad Sci U S A. 2003;100:6382–6387. [Europe PMC free article] [Abstract] [Google Scholar]
- Kish SJ, Bergeron C, Rajput A, Dozic S, Mastrogiacomo F, Chang LJ, Wilson JM, DiStefano LM, Nobrega JN. Brain cytochrome oxidase in Alzheimer’s disease. J Neurochem. 1992;59:776–779. [Abstract] [Google Scholar]
- Kiyota T, Okuyama S, Swan RJ, Jacobsen MT, Gendelman HE, Ikezu T. CNS expression of anti-inflammatory cytokine interleukin-4 attenuates Alzheimer’s disease-like pathogenesis in APP+PS1 bigenic mice. FASEB J. 2010;24:3093–3102. [Europe PMC free article] [Abstract] [Google Scholar]
- Kiyota T, Ingraham KL, Swan RJ, Jacobsen MT, Andrews SJ, Ikezu T. AAV serotype 2/1-mediated gene delivery of anti-inflammatory interleukin-10 enhances neurogenesis and cognitive function in APP+PS1 mice. Gene Ther. 2011 [Europe PMC free article] [Abstract] [Google Scholar]
- Kiyota T, Yamamoto M, Xiong H, Lambert MP, Klein WL, Gendelman HE, Ransohoff RM, Ikezu T. CCL2 accelerates microglia-mediated Abeta oligomer formation and progression of neurocognitive dysfunction. PloS one. 2009a;4:e6197. [Europe PMC free article] [Abstract] [Google Scholar]
- Kiyota T, Yamamoto M, Schroder B, Jacobsen MT, Swan RJ, Lambert MP, Klein WL, Gendelman HE, Ransohoff RM, Ikezu T. AAV1/2-mediated CNS gene delivery of dominant-negative CCL2 mutant suppresses gliosis, beta-amyloidosis, and learning impairment of APP/PS1 mice. Mol Ther. 2009b;17:803–809. [Europe PMC free article] [Abstract] [Google Scholar]
- Koning N, van Eijk M, Pouwels W, Brouwer MS, Voehringer D, Huitinga I, Hoek RM, Raes G, Hamann J. Expression of the inhibitory CD200 receptor is associated with alternative macrophage activation. J Innate Immun. 2010;2:195–200. [Abstract] [Google Scholar]
- Kopan R, Schroeter EH, Weintraub H, Nye JS. Signal transduction by activated mNotch: importance of proteolytic processing and its regulation by the extracellular domain. Proc Natl Acad Sci U S A. 1996;93:1683–1688. [Europe PMC free article] [Abstract] [Google Scholar]
- Kopydlowski KM, Salkowski CA, Cody MJ, van Rooijen N, Major J, Hamilton TA, Vogel SN. Regulation of macrophage chemokine expression by lipopolysaccharide in vitro and in vivo. J Immunol. 1999;163:1537–1544. [Abstract] [Google Scholar]
- Kretz-Rommel A, Qin F, Dakappagari N, Ravey EP, McWhirter J, Oltean D, Frederickson S, Maruyama T, Wild MA, Nolan MJ, Wu D, Springhorn J, Bowdish KS. CD200 expression on tumor cells suppresses antitumor immunity: new approaches to cancer immunotherapy. J Immunol. 2007;178:5595–5605. [Abstract] [Google Scholar]
- Kummer MP, Hermes M, Delekarte A, Hammerschmidt T, Kumar S, Terwel D, Walter J, Pape HC, Konig S, Roeber S, Jessen F, Klockgether T, Korte M, Heneka MT. Nitration of tyrosine 10 critically enhances amyloid beta aggregation and plaque formation. Neuron. 2011;71:833–844. [Abstract] [Google Scholar]
- Lambert MP, Barlow AK, Chromy BA, Edwards C, Freed R, Liosatos M, Morgan TE, Rozovsky I, Trommer B, Viola KL, Wals P, Zhang C, Finch CE, Krafft GA, Klein WL. Diffusible, nonfibrillar ligands derived from Abeta1-42 are potent central nervous system neurotoxins. Proc Natl Acad Sci U S A. 1998;95:6448–6453. [Europe PMC free article] [Abstract] [Google Scholar]
- Lee CK, Weindruch R, Prolla TA. Gene-expression profile of the ageing brain in mice. Nature genetics. 2000;25:294–297. [Abstract] [Google Scholar]
- Leissring MA, Murphy MP, Mead TR, Akbari Y, Sugarman MC, Jannatipour M, Anliker B, Muller U, Saftig P, De Strooper B, Wolfe MS, Golde TE, LaFerla FM. A physiologic signaling role for the gamma - secretase-derived intracellular fragment of APP. Proc Natl Acad Sci U S A. 2002;99:4697–4702. [Europe PMC free article] [Abstract] [Google Scholar]
- Lemere CA, Blusztajn JK, Yamaguchi H, Wisniewski T, Saido TC, Selkoe DJ. Sequence of deposition of heterogeneous amyloid beta-peptides and APO E in Down syndrome: implications for initial events in amyloid plaque formation. Neurobiology of disease. 1996;3:16–32. [Abstract] [Google Scholar]
- Levy-Lahad E, Wasco W, Poorkaj P, Romano DM, Oshima J, Pettingell WH, Yu CE, Jondro PD, Schmidt SD, Wang K, et al. Candidate gene for the chromosome 1 familial Alzheimer’s disease locus. Science. 1995;269:973–977. [Abstract] [Google Scholar]
- Li H, Hu J, Ma L, Yuan Z, Wang Y, Wang X, Xing D, Lei F, Du L. Comprehensive study of baicalin down-regulating NOD2 receptor expression of neurons with oxygen-glucose deprivation in vitro and cerebral ischemia-reperfusion in vivo. Eur J Pharmacol. 2010;649:92–99. [Abstract] [Google Scholar]
- Loerch PM, Lu T, Dakin KA, Vann JM, Isaacs A, Geula C, Wang J, Pan Y, Gabuzda DH, Li C, Prolla TA, Yankner BA. Evolution of the aging brain transcriptome and synaptic regulation. PloS one. 2008;3:e3329. [Europe PMC free article] [Abstract] [Google Scholar]
- Lotz M, Ebert S, Esselmann H, Iliev AI, Prinz M, Wiazewicz N, Wiltfang J, Gerber J, Nau R. Amyloid beta peptide 1-40 enhances the action of Toll-like receptor-2 and -4 agonists but antagonizes Toll-like receptor-9-induced inflammation in primary mouse microglial cell cultures. J Neurochem. 2005;94:289–298. [Abstract] [Google Scholar]
- Lu T, Pan Y, Kao SY, Li C, Kohane I, Chan J, Yankner BA. Gene regulation and DNA damage in the ageing human brain. Nature. 2004;429:883–891. [Abstract] [Google Scholar]
- Lue LF, Kuo YM, Roher AE, Brachova L, Shen Y, Sue L, Beach T, Kurth JH, Rydel RE, Rogers J. Soluble amyloid beta peptide concentration as a predictor of synaptic change in Alzheimer’s disease. Am J Pathol. 1999;155:853–862. [Europe PMC free article] [Abstract] [Google Scholar]
- Lund S, Christensen KV, Hedtjarn M, Mortensen AL, Hagberg H, Falsig J, Hasseldam H, Schrattenholz A, Porzgen P, Leist M. The dynamics of the LPS triggered inflammatory response of murine microglia under different culture and in vivo conditions. J Neuroimmunol. 2006;180:71–87. [Abstract] [Google Scholar]
- Lynch AM, Walsh C, Delaney A, Nolan Y, Campbell VA, Lynch MA. Lipopolysaccharide-induced increase in signalling in hippocampus is abrogated by IL-10--a role for IL-1 beta? J Neurochem. 2004;88:635–646. [Abstract] [Google Scholar]
- Lynch MA. Age-related neuroinflammatory changes negatively impact on neuronal function. Front Aging Neurosci. 2010;1:6. [Europe PMC free article] [Abstract] [Google Scholar]
- Lyons A, Griffin RJ, Costelloe CE, Clarke RM, Lynch MA. IL-4 attenuates the neuroinflammation induced by amyloid-beta in vivo and in vitro. J Neurochem. 2007a;101:771–781. [Abstract] [Google Scholar]
- Lyons A, Downer EJ, Crotty S, Nolan YM, Mills KH, Lynch MA. CD200 ligand receptor interaction modulates microglial activation in vivo and in vitro: a role for IL-4. J Neurosci. 2007b;27:8309–8313. [Europe PMC free article] [Abstract] [Google Scholar]
- Lyons A, McQuillan K, Deighan BF, O’Reilly JA, Downer EJ, Murphy AC, Watson M, Piazza A, O’Connell F, Griffin R, Mills KH, Lynch MA. Decreased neuronal CD200 expression in IL-4-deficient mice results in increased neuroinflammation in response to lipopolysaccharide. Brain Behav Immun. 2009;23:1020–1027. [Abstract] [Google Scholar]
- Mackaness GB. The monocyte in cellular immunity. Semin Hematol. 1970;7:172–184. [Abstract] [Google Scholar]
- Maeda A, Sobel RA. Matrix metalloproteinases in the normal human central nervous system, microglial nodules, and multiple sclerosis lesions. J Neuropathol Exp Neurol. 1996;55:300–309. [Abstract] [Google Scholar]
- Magavi SS, Mitchell BD, Szentirmai O, Carter BS, Macklis JD. Adult-born and preexisting olfactory granule neurons undergo distinct experience-dependent modifications of their olfactory responses in vivo. J Neurosci. 2005;25:10729–10739. [Europe PMC free article] [Abstract] [Google Scholar]
- Maher FO, Nolan Y, Lynch MA. Downregulation of IL-4-induced signalling in hippocampus contributes to deficits in LTP in the aged rat. Neurobiol Aging. 2005;26:717–728. [Abstract] [Google Scholar]
- Maher FO, Clarke RM, Kelly A, Nally RE, Lynch MA. Interaction between interferon gamma and insulin-like growth factor-1 in hippocampus impacts on the ability of rats to sustain long-term potentiation. J Neurochem. 2006;96:1560–1571. [Abstract] [Google Scholar]
- Mantovani A, Sica A, Sozzani S, Allavena P, Vecchi A, Locati M. The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol. 2004;25:677–686. [Abstract] [Google Scholar]
- Martin BK, Szekely C, Brandt J, Piantadosi S, Breitner JC, Craft S, Evans D, Green R, Mullan M. Cognitive function over time in the Alzheimer’s Disease Anti-inflammatory Prevention Trial (ADAPT): results of a randomized, controlled trial of naproxen and celecoxib. Archives of neurology. 2008;65:896–905. [Europe PMC free article] [Abstract] [Google Scholar]
- Matsuda H. The role of neuroimaging in mild cognitive impairment. Neuropathology. 2007;27:570–577. [Abstract] [Google Scholar]
- Mattson MP, Partin J, Begley JG. Amyloid beta-peptide induces apoptosis-related events in synapses and dendrites. Brain Res. 1998;807:167–176. [Abstract] [Google Scholar]
- McGeer EG, McGeer PL. Neuroinflammation in Alzheimer’s disease and mild cognitive impairment: a field in its infancy. J Alzheimers Dis. 2010;19:355–361. [Abstract] [Google Scholar]
- McGeer PL, Rogers J, McGeer EG. Neuroimmune mechanisms in Alzheimer disease pathogenesis. Alzheimer Dis Assoc Disord. 1994;8:149–158. [Abstract] [Google Scholar]
- McGeer PL, Akiyama H, Itagaki S, McGeer EG. Immune system response in Alzheimer’s disease. Can J Neurol Sci. 1989;16:516–527. [Abstract] [Google Scholar]
- McMaster WR, Williams AF. Identification of Ia glycoproteins in rat thymus and purification from rat spleen. Eur J Immunol. 1979;9:426–433. [Abstract] [Google Scholar]
- Mihrshahi R, Barclay AN, Brown MH. Essential roles for Dok2 and RasGAP in CD200 receptor-mediated regulation of human myeloid cells. J Immunol. 2009;183:4879–4886. [Europe PMC free article] [Abstract] [Google Scholar]
- Mills CD, Kincaid K, Alt JM, Heilman MJ, Hill AM. M-1/M-2 macrophages and the Th1/Th2 paradigm. J Immunol. 2000;164:6166–6173. [Abstract] [Google Scholar]
- Minati L, Edginton T, Bruzzone MG, Giaccone G. Current concepts in Alzheimer’s disease: a multidisciplinary review. Am J Alzheimers Dis Other Demen. 2009;24:95–121. [Europe PMC free article] [Abstract] [Google Scholar]
- Ming GL, Song H. Adult neurogenesis in the mammalian central nervous system. Annu Rev Neurosci. 2005;28:223–250. [Abstract] [Google Scholar]
- Monje ML, Toda H, Palmer TD. Inflammatory blockade restores adult hippocampal neurogenesis. Science. 2003;302:1760–1765. [Abstract] [Google Scholar]
- Moreaux J, Veyrune JL, Reme T, De Vos J, Klein B. CD200: a putative therapeutic target in cancer. Biochem Biophys Res Commun. 2008;366:117–122. [Abstract] [Google Scholar]
- Moreaux J, Hose D, Reme T, Jourdan E, Hundemer M, Legouffe E, Moine P, Bourin P, Moos M, Corre J, Mohler T, De Vos J, Rossi JF, Goldschmidt H, Klein B. CD200 is a new prognostic factor in multiple myeloma. Blood. 2006;108:4194–4197. [Abstract] [Google Scholar]
- Nagai Y, Akashi S, Nagafuku M, Ogata M, Iwakura Y, Akira S, Kitamura T, Kosugi A, Kimoto M, Miyake K. Essential role of MD-2 in LPS responsiveness and TLR4 distribution. Nat Immunol. 2002;3:667–672. [Abstract] [Google Scholar]
- Niederkorn JY. See no evil, hear no evil, do no evil: the lessons of immune privilege. Nat Immunol. 2006;7:354–359. [Abstract] [Google Scholar]
- Nolan Y, Maher FO, Martin DS, Clarke RM, Brady MT, Bolton AE, Mills KH, Lynch MA. Role of interleukin-4 in regulation of age-related inflammatory changes in the hippocampus. J Biol Chem. 2005;280:9354–9362. [Abstract] [Google Scholar]
- Oitzl MS, van Oers H, Schobitz B, de Kloet ER. Interleukin-1 beta, but not interleukin-6, impairs spatial navigation learning. Brain Res. 1993;613:160–163. [Abstract] [Google Scholar]
- Okello A, Edison P, Archer HA, Turkheimer FE, Kennedy J, Bullock R, Walker Z, Kennedy A, Fox N, Rossor M, Brooks DJ. Microglial activation and amyloid deposition in mild cognitive impairment: a PET study. Neurology. 2009;72:56–62. [Europe PMC free article] [Abstract] [Google Scholar]
- Oppenheim JJ, Rosenstreich DL. Signals regulating in vitro activation of lymphocytes. Prog Allergy. 1976;20:65–194. [Abstract] [Google Scholar]
- Patel NS, Paris D, Mathura V, Quadros AN, Crawford FC, Mullan MJ. Inflammatory cytokine levels correlate with amyloid load in transgenic mouse models of Alzheimer’s disease. J Neuroinflammation. 2005;2:9. [Europe PMC free article] [Abstract] [Google Scholar]
- Petermann KB, Rozenberg GI, Zedek D, Groben P, McKinnon K, Buehler C, Kim WY, Shields JM, Penland S, Bear JE, Thomas NE, Serody JS, Sharpless NE. CD200 is induced by ERK and is a potential therapeutic target in melanoma. J Clin Invest. 2007;117:3922–3929. [Europe PMC free article] [Abstract] [Google Scholar]
- Plunkett JA, Yu CG, Easton JM, Bethea JR, Yezierski RP. Effects of interleukin-10 (IL-10) on pain behavior and gene expression following excitotoxic spinal cord injury in the rat. Exp Neurol. 2001;168:144–154. [Abstract] [Google Scholar]
- Poewe W. Non-motor symptoms in Parkinson’s disease. Eur J Neurol. 2008;15(Suppl 1):14–20. [Abstract] [Google Scholar]
- Pratico D, Sung S. Lipid peroxidation and oxidative imbalance: early functional events in Alzheimer’s disease. J Alzheimers Dis. 2004;6:171–175. [Abstract] [Google Scholar]
- Pratico D, Uryu K, Leight S, Trojanoswki JQ, Lee VM. Increased lipid peroxidation precedes amyloid plaque formation in an animal model of Alzheimer amyloidosis. J Neurosci. 2001;21:4183–4187. [Europe PMC free article] [Abstract] [Google Scholar]
- Prolla TA. DNA microarray analysis of the aging brain. Chem Senses. 2002;27:299–306. [Abstract] [Google Scholar]
- Qin H, Wilson CA, Roberts KL, Baker BJ, Zhao X, Benveniste EN. IL-10 inhibits lipopolysaccharide-induced CD40 gene expression through induction of suppressor of cytokine signaling-3. J Immunol. 2006;177:7761–7771. [Abstract] [Google Scholar]
- Raes G, De Baetselier P, Noel W, Beschin A, Brombacher F, Hassanzadeh Gh G. Differential expression of FIZZ1 and Ym1 in alternatively versus classically activated macrophages. J Leukoc Biol. 2002;71:597–602. [Abstract] [Google Scholar]
- Ravetch JV, Kinet JP. Fc receptors. Annu Rev Immunol. 1991;9:457–492. [Abstract] [Google Scholar]
- Ravetch JV, Lanier LL. Immune inhibitory receptors. Science. 2000;290:84–89. [Abstract] [Google Scholar]
- Reed-Geaghan EG, Savage JC, Hise AG, Landreth GE. CD14 and toll-like receptors 2 and 4 are required for fibrillar A{beta}-stimulated microglial activation. J Neurosci. 2009;29:11982–11992. [Europe PMC free article] [Abstract] [Google Scholar]
- Reed-Geaghan EG, Reed QW, Cramer PE, Landreth GE. Deletion of CD14 attenuates Alzheimer’s disease pathology by influencing the brain’s inflammatory milieu. J Neurosci. 2010;30:15369–15373. [Europe PMC free article] [Abstract] [Google Scholar]
- Rosenberg GA. Matrix metalloproteinases and their multiple roles in neurodegenerative diseases. Lancet Neurol. 2009;8:205–216. [Abstract] [Google Scholar]
- Rosenberg GA, Sullivan N, Esiri MM. White matter damage is associated with matrix metalloproteinases in vascular dementia. Stroke. 2001;32:1162–1168. [Abstract] [Google Scholar]
- Salloway S, Sperling R, Gilman S, Fox NC, Blennow K, Raskind M, Sabbagh M, Honig LS, Doody R, van Dyck CH, Mulnard R, Barakos J, Gregg KM, Liu E, Lieberburg I, Schenk D, Black R, Grundman M. A phase 2 multiple ascending dose trial of bapineuzumab in mild to moderate Alzheimer disease. Neurology. 2009;73:2061–2070. [Europe PMC free article] [Abstract] [Google Scholar]
- Samavati L, Lee I, Mathes I, Lottspeich F, Huttemann M. Tumor necrosis factor alpha inhibits oxidative phosphorylation through tyrosine phosphorylation at subunit I of cytochrome c oxidase. J Biol Chem. 2008;283:21134–21144. [Europe PMC free article] [Abstract] [Google Scholar]
- Scheff SW, Price DA, Schmitt FA, Mufson EJ. Hippocampal synaptic loss in early Alzheimer’s disease and mild cognitive impairment. Neurobiol Aging. 2006;27:1372–1384. [Abstract] [Google Scholar]
- Scheff SW, Price DA, Schmitt FA, DeKosky ST, Mufson EJ. Synaptic alterations in CA1 in mild Alzheimer disease and mild cognitive impairment. Neurology. 2007;68:1501–1508. [Abstract] [Google Scholar]
- Schroeter EH, Kisslinger JA, Kopan R. Notch-1 signalling requires ligand-induced proteolytic release of intracellular domain. Nature. 1998;393:382–386. [Abstract] [Google Scholar]
- Selkoe DJ. The molecular pathology of Alzheimer’s disease. Neuron. 1991;6:487–498. [Abstract] [Google Scholar]
- Selkoe DJ. Amyloid beta-protein and the genetics of Alzheimer’s disease. J Biol Chem. 1996;271:18295–18298. [Abstract] [Google Scholar]
- Serrats J, Schiltz JC, Garcia-Bueno B, van Rooijen N, Reyes TM, Sawchenko PE. Dual roles for perivascular macrophages in immune-to-brain signaling. Neuron. 2010;65:94–106. [Europe PMC free article] [Abstract] [Google Scholar]
- Seubert P, Vigo-Pelfrey C, Esch F, Lee M, Dovey H, Davis D, Sinha S, Schlossmacher M, Whaley J, Swindlehurst C, et al. Isolation and quantification of soluble Alzheimer’s beta-peptide from biological fluids. Nature. 1992;359:325–327. [Abstract] [Google Scholar]
- Seubert P, Oltersdorf T, Lee MG, Barbour R, Blomquist C, Davis DL, Bryant K, Fritz LC, Galasko D, Thal LJ, et al. Secretion of beta-amyloid precursor protein cleaved at the amino terminus of the beta-amyloid peptide. Nature. 1993;361:260–263. [Abstract] [Google Scholar]
- Shaftel SS, Kyrkanides S, Olschowka JA, Miller JN, Johnson RE, O’Banion MK. Sustained hippocampal IL-1 beta overexpression mediates chronic neuroinflammation and ameliorates Alzheimer plaque pathology. J Clin Invest. 2007;117:1595–1604. [Europe PMC free article] [Abstract] [Google Scholar]
- Shankar GM, Walsh DM. Alzheimer’s disease: synaptic dysfunction and Abeta. Mol Neurodegener. 2009;4:48. [Europe PMC free article] [Abstract] [Google Scholar]
- Shein NA, Grigoriadis N, Horowitz M, Umschwief G, Alexandrovich AG, Simeonidou C, Grigoriadis S, Touloumi O, Shohami E. Microglial involvement in neuroprotection following experimental traumatic brain injury in heat-acclimated mice. Brain Res. 2008;1244:132–141. [Abstract] [Google Scholar]
- Sherrington R, Rogaev EI, Liang Y, Rogaeva EA, Levesque G, Ikeda M, Chi H, Lin C, Li G, Holman K, Tsuda T, Mar L, Foncin JF, Bruni AC, Montesi MP, Sorbi S, Rainero I, Pinessi L, Nee L, Chumakov I, Pollen D, Brookes A, Sanseau P, Polinsky RJ, Wasco W, Da Silva HA, Haines JL, Perkicak-Vance MA, Tanzi RE, Roses AD, Fraser PE, Rommens JM, St George-Hyslop PH. Cloning of a gene bearing missense mutations in early-onset familial Alzheimer’s disease. Nature. 1995;375:754–760. [Abstract] [Google Scholar]
- Shinohara H, Inoue A, Toyama-Sorimachi N, Nagai Y, Yasuda T, Suzuki H, Horai R, Iwakura Y, Yamamoto T, Karasuyama H, Miyake K, Yamanashi Y. Dok-1 and Dok-2 are negative regulators of lipopolysaccharide-induced signaling. J Exp Med. 2005;201:333–339. [Europe PMC free article] [Abstract] [Google Scholar]
- Shiryaev SA, Remacle AG, Savinov AY, Chernov AV, Cieplak P, Radichev IA, Williams R, Shiryaeva TN, Gawlik K, Postnova TI, Ratnikov BI, Eroshkin AM, Motamedchaboki K, Smith JW, Strongin AY. Inflammatory proprotein convertase-matrix metalloproteinase proteolytic pathway in antigen-presenting cells as a step to autoimmune multiple sclerosis. J Biol Chem. 2009;284:30615–30626. [Europe PMC free article] [Abstract] [Google Scholar]
- Shoji M, Golde TE, Ghiso J, Cheung TT, Estus S, Shaffer LM, Cai XD, McKay DM, Tintner R, Frangione B, et al. Production of the Alzheimer amyloid beta protein by normal proteolytic processing. Science. 1992;258:126–129. [Abstract] [Google Scholar]
- Sierra A, Encinas JM, Deudero JJ, Chancey JH, Enikolopov G, Overstreet-Wadiche LS, Tsirka SE, Maletic-Savatic M. Microglia shape adult hippocampal neurogenesis through apoptosis-coupled phagocytosis. Cell Stem Cell. 2010;7:483–495. [Europe PMC free article] [Abstract] [Google Scholar]
- Siva A, Xin H, Qin F, Oltean D, Bowdish KS, Kretz-Rommel A. Immune modulation by melanoma and ovarian tumor cells through expression of the immunosuppressive molecule CD200. Cancer Immunol Immunother. 2008;57:987–996. [Europe PMC free article] [Abstract] [Google Scholar]
- Snelgrove RJ, Goulding J, Didierlaurent AM, Lyonga D, Vekaria S, Edwards L, Gwyer E, Sedgwick JD, Barclay AN, Hussell T. A critical function for CD200 in lung immune homeostasis and the severity of influenza infection. Nat Immunol. 2008;9:1074–1083. [Abstract] [Google Scholar]
- So K, Moriya T, Nishitani S, Takahashi H, Shinohara K. The olfactory conditioning in the early postnatal period stimulated neural stem/progenitor cells in the subventricular zone and increased neurogenesis in the olfactory bulb of rats. Neuroscience. 2008;151:120–128. [Abstract] [Google Scholar]
- Song M, Jin J, Lim JE, Kou J, Pattanayak A, Rehman JA, Kim HD, Tahara K, Lalonde R, Fukuchi K. TLR4 mutation reduces microglial activation, increases Abeta deposits and exacerbates cognitive deficits in a mouse model of Alzheimer’s disease. J Neuroinflammation. 2011;8:92. [Europe PMC free article] [Abstract] [Google Scholar]
- Sparkman NL, Johnson RW. Neuroinflammation associated with aging sensitizes the brain to the effects of infection or stress. Neuroimmunomodulation. 2008;15:323–330. [Europe PMC free article] [Abstract] [Google Scholar]
- Stadler J, Bentz BG, Harbrecht BG, Di Silvio M, Curran RD, Billiar TR, Hoffman RA, Simmons RL. Tumor necrosis factor alpha inhibits hepatocyte mitochondrial respiration. Ann Surg. 1992;216:539–546. [Abstract] [Google Scholar]
- Struble RG, Ala T, Patrylo PR, Brewer GJ, Yan XX. Is brain amyloid production a cause or a result of dementia of the Alzheimer’s type? J Alzheimers Dis. 2010;22:393–399. [Europe PMC free article] [Abstract] [Google Scholar]
- Tahara K, Kim HD, Jin JJ, Maxwell JA, Li L, Fukuchi K. Role of toll-like receptor signalling in Abeta uptake and clearance. Brain: a journal of neurology. 2006;129:3006–3019. [Europe PMC free article] [Abstract] [Google Scholar]
- Takasugi N, Tomita T, Hayashi I, Tsuruoka M, Niimura M, Takahashi Y, Thinakaran G, Iwatsubo T. The role of presenilin cofactors in the gamma-secretase complex. Nature. 2003;422:438–441. [Abstract] [Google Scholar]
- Takeda K, Akira S. Microbial recognition by Toll-like receptors. J Dermatol Sci. 2004;34:73–82. [Abstract] [Google Scholar]
- Tamura Y, Kataoka Y, Cui Y, Takamori Y, Watanabe Y, Yamada H. Multi-directional differentiation of doublecortin- and NG2-immunopositive progenitor cells in the adult rat neocortex in vivo. Eur J Neurosci. 2007;25:3489–3498. [Abstract] [Google Scholar]
- Tang SC, Arumugam TV, Xu X, Cheng A, Mughal MR, Jo DG, Lathia JD, Siler DA, Chigurupati S, Ouyang X, Magnus T, Camandola S, Mattson MP. Pivotal role for neuronal Toll-like receptors in ischemic brain injury and functional deficits. Proc Natl Acad Sci U S A. 2007;104:13798–13803. [Europe PMC free article] [Abstract] [Google Scholar]
- Tesseur I, Zou K, Esposito L, Bard F, Berber E, Can JV, Lin AH, Crews L, Tremblay P, Mathews P, Mucke L, Masliah E, Wyss-Coray T. Deficiency in neuronal TGF-beta signaling promotes neurodegeneration and Alzheimer’s pathology. J Clin Invest. 2006;116:3060–3069. [Europe PMC free article] [Abstract] [Google Scholar]
- Town T, Laouar Y, Pittenger C, Mori T, Szekely CA, Tan J, Duman RS, Flavell RA. Blocking TGF-beta-Smad2/3 innate immune signaling mitigates Alzheimer-like pathology. Nature medicine. 2008;14:681–687. [Europe PMC free article] [Abstract] [Google Scholar]
- Tronel S, Belnoue L, Grosjean N, Revest JM, Piazza PV, Koehl M, Abrous DN. Adult-born neurons are necessary for extended contextual discrimination. Hippocampus. 2010 [Abstract] [Google Scholar]
- Unkeless JC, Scigliano E, Freedman VH. Structure and function of human and murine receptors for IgG. Annu Rev Immunol. 1988;6:251–281. [Abstract] [Google Scholar]
- Vassar R, Bennett BD, Babu-Khan S, Kahn S, Mendiaz EA, Denis P, Teplow DB, Ross S, Amarante P, Loeloff R, Luo Y, Fisher S, Fuller J, Edenson S, Lile J, Jarosinski MA, Biere AL, Curran E, Burgess T, Louis JC, Collins F, Treanor J, Rogers G, Citron M. Beta-secretase cleavage of Alzheimer’s amyloid precursor protein by the transmembrane aspartic protease BACE. Science. 1999;286:735–741. [Abstract] [Google Scholar]
- Villeda SA, Luo J, Mosher KI, Zou B, Britschgi M, Bieri G, Stan TM, Fainberg N, Ding Z, Eggel A, Lucin KM, Czirr E, Park JS, Couillard-Despres S, Aigner L, Li G, Peskind ER, Kaye JA, Quinn JF, Galasko DR, Xie XS, Rando TA, Wyss-Coray T. The ageing systemic milieu negatively regulates neurogenesis and cognitive function. Nature. 2011;477:90–94. [Europe PMC free article] [Abstract] [Google Scholar]
- Voloboueva LA, Giffard RG. Inflammation, mitochondria, and the inhibition of adult neurogenesis. J Neurosci Res. 2011 [Europe PMC free article] [Abstract] [Google Scholar]
- Waldau B, Shetty AK. Behavior of neural stem cells in the Alzheimer brain. Cell Mol Life Sci. 2008;65:2372–2384. [Europe PMC free article] [Abstract] [Google Scholar]
- Walker DG, Dalsing-Hernandez JE, Campbell NA, Lue LF. Decreased expression of CD200 and CD200 receptor in Alzheimer’s disease: a potential mechanism leading to chronic inflammation. Exp Neurol. 2009;215:5–19. [Europe PMC free article] [Abstract] [Google Scholar]
- Walsh DM, Klyubin I, Fadeeva JV, Cullen WK, Anwyl R, Wolfe MS, Rowan MJ, Selkoe DJ. Naturally secreted oligomers of amyloid beta protein potently inhibit hippocampal long-term potentiation in vivo. Nature. 2002;416:535–539. [Abstract] [Google Scholar]
- Walter J, Honsek SD, Illes S, Wellen JM, Hartung HP, Rose CR, Dihne M. A new role for interferon gamma in neural stem/precursor cell dysregulation. Mol Neurodegener. 2011;6:18. [Europe PMC free article] [Abstract] [Google Scholar]
- Webb M, Barclay AN. Localisation of the MRC OX-2 glycoprotein on the surfaces of neurones. J Neurochem. 1984;43:1061–1067. [Abstract] [Google Scholar]
- Wegiel J, Wisniewski HM. The complex of microglial cells and amyloid star in three-dimensional reconstruction. Acta Neuropathol. 1990;81:116–124. [Abstract] [Google Scholar]
- Wen PH, Hof PR, Chen X, Gluck K, Austin G, Younkin SG, Younkin LH, DeGasperi R, Gama Sosa MA, Robakis NK, Haroutunian V, Elder GA. The presenilin-1 familial Alzheimer disease mutant P117L impairs neurogenesis in the hippocampus of adult mice. Exp Neurol. 2004;188:224–237. [Abstract] [Google Scholar]
- Wilcock DM, Lewis MR, Van Nostrand WE, Davis J, Previti ML, Gharkholonarehe N, Vitek MP, Colton CA. Progression of amyloid pathology to Alzheimer’s disease pathology in an amyloid precursor protein transgenic mouse model by removal of nitric oxide synthase 2. J Neurosci. 2008;28:1537–1545. [Europe PMC free article] [Abstract] [Google Scholar]
- Wiley CA, Lopresti BJ, Venneti S, Price J, Klunk WE, DeKosky ST, Mathis CA. Carbon 11-labeled Pittsburgh Compound B and carbon 11-labeled (R)-PK11195 positron emission tomographic imaging in Alzheimer disease. Archives of neurology. 2009;66:60–67. [Europe PMC free article] [Abstract] [Google Scholar]
- Wisniewski HM, Wegiel J. Spatial relationships between astrocytes and classical plaque components. Neurobiol Aging. 1991;12:593–600. [Abstract] [Google Scholar]
- Wright GJ, Puklavec MJ, Willis AC, Hoek RM, Sedgwick JD, Brown MH, Barclay AN. Lymphoid/neuronal cell surface OX2 glycoprotein recognizes a novel receptor on macrophages implicated in the control of their function. Immunity. 2000;13:233–242. [Abstract] [Google Scholar]
- Wright GJ, Cherwinski H, Foster-Cuevas M, Brooke G, Puklavec MJ, Bigler M, Song Y, Jenmalm M, Gorman D, McClanahan T, Liu MR, Brown MH, Sedgwick JD, Phillips JH, Barclay AN. Characterization of the CD200 receptor family in mice and humans and their interactions with CD200. J Immunol. 2003;171:3034–3046. [Abstract] [Google Scholar]
- Wyss-Coray T, Yan F, Lin AH, Lambris JD, Alexander JJ, Quigg RJ, Masliah E. Prominent neurodegeneration and increased plaque formation in complement-inhibited Alzheimer’s mice. Proc Natl Acad Sci U S A. 2002;99:10837–10842. [Europe PMC free article] [Abstract] [Google Scholar]
- Wyss-Coray T, Lin C, Yan F, Yu GQ, Rohde M, McConlogue L, Masliah E, Mucke L. TGF-beta1 promotes microglial amyloid-beta clearance and reduces plaque burden in transgenic mice. Nature medicine. 2001;7:612–618. [Abstract] [Google Scholar]
- Xapelli S, Bernardino L, Ferreira R, Grade S, Silva AP, Salgado JR, Cavadas C, Grouzmann E, Poulsen FR, Jakobsen B, Oliveira CR, Zimmer J, Malva JO. Interaction between neuropeptide Y (NPY) and brain-derived neurotrophic factor in NPY-mediated neuroprotection against excitotoxicity: a role for microglia. Eur J Neurosci. 2008;27:2089–2102. [Abstract] [Google Scholar]
- Yamamoto M, Kiyota T, Walsh SM, Liu J, Kipnis J, Ikezu T. Cytokine-mediated inhibition of fibrillar amyloid-beta peptide degradation by human mononuclear phagocytes. J Immunol. 2008;181:3877–3886. [Europe PMC free article] [Abstract] [Google Scholar]
- Yamamoto M, Kiyota T, Horiba M, Buescher JL, Walsh SM, Gendelman HE, Ikezu T. Interferon-gamma and tumor necrosis factor-alpha regulate amyloid-beta plaque deposition and beta-secretase expression in Swedish mutant APP transgenic mice. Am J Pathol. 2007;170:680–692. [Europe PMC free article] [Abstract] [Google Scholar]
- Yasuda T, Bundo K, Hino A, Honda K, Inoue A, Shirakata M, Osawa M, Tamura T, Nariuchi H, Oda H, Yamamoto T, Yamanashi Y. Dok-1 and Dok-2 are negative regulators of T cell receptor signaling. Int Immunol. 2007;19:487–495. [Abstract] [Google Scholar]
- Yokokura M, Mori N, Yagi S, Yoshikawa E, Kikuchi M, Yoshihara Y, Wakuda T, Sugihara G, Takebayashi K, Suda S, Iwata Y, Ueki T, Tsuchiya KJ, Suzuki K, Nakamura K, Ouchi Y. In vivo changes in microglial activation and amyloid deposits in brain regions with hypometabolism in Alzheimer’s disease. European journal of nuclear medicine and molecular imaging. 2011;38:343–351. [Abstract] [Google Scholar]
- Zell R, Geck P, Werdan K, Boekstegers P. TNF-alpha and IL-1 alpha inhibit both pyruvate dehydrogenase activity and mitochondrial function in cardiomyocytes: evidence for primary impairment of mitochondrial function. Mol Cell Biochem. 1997;177:61–67. [Abstract] [Google Scholar]
- Zhang S, Cherwinski H, Sedgwick JD, Phillips JH. Molecular mechanisms of CD200 inhibition of mast cell activation. J Immunol. 2004;173:6786–6793. [Abstract] [Google Scholar]
- Zhang YW, Thompson R, Zhang H, Xu H. APP processing in Alzheimer’s disease. Mol Brain. 2011;4:3. [Europe PMC free article] [Abstract] [Google Scholar]
- Zhao W, Xie W, Xiao Q, Beers DR, Appel SH. Protective effects of an anti-inflammatory cytokine, interleukin-4, on motoneuron toxicity induced by activated microglia. J Neurochem. 2006;99:1176–1187. [Abstract] [Google Scholar]
Full text links
Read article at publisher's site: https://doi.org/10.1007/s00005-012-0181-2
Read article for free, from open access legal sources, via Unpaywall: https://europepmc.org/articles/pmc4429536?pdf=render
Citations & impact
Impact metrics
Article citations
Magnetic stirring with iron oxide nanospinners accretes neurotoxic Aβ42 oligomers into phagocytic clearable plaques for Alzheimer's disease treatment.
Mater Today Bio, 28:101213, 27 Aug 2024
Cited by: 0 articles | PMID: 39280110 | PMCID: PMC11402446
The intricate interplay between microglia and adult neurogenesis in Alzheimer's disease.
Front Cell Neurosci, 18:1456253, 18 Sep 2024
Cited by: 0 articles | PMID: 39360265 | PMCID: PMC11445663
Review Free full text in Europe PMC
Microglial senescence in neurodegeneration: Insights, implications, and therapeutic opportunities.
Neuroprotection, 2(3):182-195, 15 Sep 2024
Cited by: 0 articles | PMID: 39364217 | PMCID: PMC11449118
Microglia and Astrocytes in Alzheimer's Disease: Significance and Summary of Recent Advances.
Aging Dis, 15(4):1537-1564, 01 Aug 2024
Cited by: 5 articles | PMID: 37815901 | PMCID: PMC11272214
Review Free full text in Europe PMC
Single-Cell RNA Sequencing Reveals Immunomodulatory Effects of Stem Cell Factor and Granulocyte Colony-Stimulating Factor Treatment in the Brains of Aged APP/PS1 Mice.
Biomolecules, 14(7):827, 10 Jul 2024
Cited by: 0 articles | PMID: 39062541 | PMCID: PMC11275138
Go to all (217) article citations
Similar Articles
To arrive at the top five similar articles we use a word-weighted algorithm to compare words from the Title and Abstract of each citation.
Neuroinflammation and Alzheimer's Disease: Implications for Microglial Activation.
Curr Alzheimer Res, 14(11):1140-1148, 01 Jan 2017
Cited by: 87 articles | PMID: 28164764
Review
Influence of microglia and astrocyte activation in the neuroinflammatory pathogenesis of Alzheimer's disease: Rational insights for the therapeutic approaches.
J Clin Neurosci, 59:6-11, 29 Oct 2018
Cited by: 67 articles | PMID: 30385170
Review
Identification and therapeutic modulation of a pro-inflammatory subset of disease-associated-microglia in Alzheimer's disease.
Mol Neurodegener, 13(1):24, 21 May 2018
Cited by: 204 articles | PMID: 29784049 | PMCID: PMC5963076
Quantitative proteomics of acutely-isolated mouse microglia identifies novel immune Alzheimer's disease-related proteins.
Mol Neurodegener, 13(1):34, 28 Jun 2018
Cited by: 62 articles | PMID: 29954413 | PMCID: PMC6025801
Funding
Funders who supported this work.
NIA NIH HHS (2)
Grant ID: AG032600
Grant ID: R21 AG032600
NIGMS NIH HHS (2)
Grant ID: T32 GM008541
Grant ID: 5T32GM008541
NIMH NIH HHS (2)
Grant ID: MH072539
Grant ID: R01 MH072539